1
|
Zhang H, Pang Y, Yi L, Wang X, Wei P, Wang H, Lin S. Epigenetic regulators combined with tumour immunotherapy: current status and perspectives. Clin Epigenetics 2025; 17:51. [PMID: 40119465 PMCID: PMC11929245 DOI: 10.1186/s13148-025-01856-6] [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: 12/31/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
Immunotherapy, particularly immune checkpoint inhibitor therapy, has demonstrated clinical benefits in solid tumours. Despite its satisfactory clinical efficacy, it still faces several issues, such as limited eligibility, low response rates and cytotoxicity. Cancer epigenetics implies that tumour cells exhibit unique phenotypes because of their unique characteristics, thus reprogramming of the epigenome holds promise for cancer therapy. Epigenetic regulation plays an important role in regulating gene expression during tumour development and maintenance. Epigenetic regulators induce cancer cell cycle arrest, apoptosis and differentiation of cancer cells, thereby exerting anti-tumour effects. Recent studies have revealed a significant correlation between epigenetic regulatory factors and immune checkpoint therapy. Epigenetics can modulate various aspects of the tumour immune microenvironment and immune response to enhance the sensitivity of immunotherapy, such as lowering the concentration required and mitigating cytotoxicity. This review primarily discusses DNA methyltransferase inhibitors, histone deacetylase inhibitors, enhancer of zeste homolog 2 inhibitors and lysine-specific demethylase 1 inhibitors, which are associated with transcriptional repression. This repression alters the expression of genes involved in the immune checkpoint, thereby enhancing the effectiveness of immunotherapy. We also discuss the potential and challenges of tumour immunotherapy and highlight its advantages, application challenges and clinical research on integrating epigenetic regulatory factors with tumour immunotherapy.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Gastroenterology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Yutong Pang
- Department of Gastroenterology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Ling Yi
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Xiaojue Wang
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Panjian Wei
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Haichao Wang
- Institute of Resources and Environment, Beijing Academy of Science and Technology, Beijing, 100089, China.
| | - Shuye Lin
- Department of Gastroenterology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
2
|
Vlastou E, Kougioumtzopoulou A, Platoni K, Georgakopoulos I, Lagopati N, Kouloulias V, Zygogianni A. The Emerging Role of Nanoparticles Combined with Either Radiotherapy or Hyperthermia in Head and Neck Cancer: A Current Review. Cancers (Basel) 2025; 17:899. [PMID: 40075746 PMCID: PMC11899074 DOI: 10.3390/cancers17050899] [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: 01/16/2025] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Head and neck cancer (HNC) includes various malignancies and represents the seventh most common cancer worldwide. The early diagnosis of HNC results in a 70-90% five-year survival rate, which declines with locally advanced stages of disease. Current care employs a multimodal strategy encompassing surgery, radiation therapy (RT), chemotherapy, and immunotherapy, while treatment options vary according to the stage, tumor features, and patient characteristics. About 75% of patients with HNC will benefit from RT, either as a primary treatment or as adjuvant therapy following surgical resection. Technological improvements in RT, such as intensity-modulated RT (IMRT) and image-guided RT (IGRT), have enhanced tumor targeting and minimized adjacent healthy tissue irradiation while also expanding RT to the recurrent or metastatic setting. Innovative therapeutic strategies for HNC integrate RT with immunotherapy, gene therapy, molecular targeted therapy, photodynamic therapy, photothermal therapy, and nanoparticles (NPs), with the objective of optimizing tumor control while reducing damage to normal tissues. NPs are emerging as possible radiosensitizers in HNC treatment, enhancing the efficacy of RT, chemotherapy, and immunotherapy. In vivo and in vitro studies on the irradiation of tumors containing gold (Au), gadolinium (Gd), and hafnium oxide (HfO2) NPs show promising results in enhancing tumor destruction and survival rates, indicating their potential for clinical application. Hyperthermia, investigated as an adjunct treatment, potentially improves outcomes when combined with RT or chemotherapy, with advancements in nanotechnology renewing interest in this approach in HNC. At present, NBTXR3 is the sole NP that is being investigated in clinical trials for the enhancement of HNC RT.
Collapse
Affiliation(s)
- Elena Vlastou
- Radiotherapy Department, General Children’s Hospital ‘Pan. & Aglaia Kyriakou’, 11527 Athens, Greece;
| | - Andromachi Kougioumtzopoulou
- Department of Clinical Radiation Oncology, “ATTIKON” General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Haidari, Greece; (A.K.); (K.P.)
| | - Kalliopi Platoni
- Department of Clinical Radiation Oncology, “ATTIKON” General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Haidari, Greece; (A.K.); (K.P.)
| | - Ioannis Georgakopoulos
- Radiotherapy Unit, 1st Radiology Department, ‘Aretaieion’ University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.G.); (A.Z.)
| | - Nefeli Lagopati
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vasileios Kouloulias
- Department of Clinical Radiation Oncology, “ATTIKON” General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Haidari, Greece; (A.K.); (K.P.)
| | - Anna Zygogianni
- Radiotherapy Unit, 1st Radiology Department, ‘Aretaieion’ University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece; (I.G.); (A.Z.)
| |
Collapse
|
3
|
Abreu MM, Chocron AF, Smadja DM. From cold to hot: mechanisms of hyperthermia in modulating tumor immunology for enhanced immunotherapy. Front Immunol 2025; 16:1487296. [PMID: 40092992 PMCID: PMC11906415 DOI: 10.3389/fimmu.2025.1487296] [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: 08/27/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
The emergence of immunotherapies has revolutionized cancer treatment by leveraging the immune system to target malignancies, offering new hope where traditional therapies often fall short. Within this context, hyperthermia (HT) has re-emerged as a promising adjunctive treatment, capable of enhancing the effectiveness of radiotherapy, chemotherapy, and immunotherapy. HT influences both the innate and adaptive immune systems, enhancing the activity of immune cells such as neutrophils, NK cells, and dendritic cells, while also modulating the tumor microenvironment (TME) to promote immunogenic cell death (ICD) and reduce immunosuppressive conditions. These effects contribute to the transformation of immunologically "cold" tumors into "hot" tumors, making them more susceptible to immune-mediated destruction. Furthermore, HT can amplify the efficacy of immune checkpoint inhibitors (ICIs) by improving immune cell infiltration, inducing damage-associated molecular pattern (DAMP) release, and enhancing antigen presentation. Preclinical and clinical studies support the combination of HT with ICIs, demonstrating improved outcomes in otherwise resistant tumors. However, the full therapeutic potential of the different technologies allowing to apply HT remains to be fully understood, and further research is needed to optimize treatment protocols, explore the differential impacts of local versus whole-body hyperthermia, and identify biomarkers for patient stratification. This review underscores the multifaceted role of HT in immunity and its potential to significantly enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- M Marc Abreu
- Medicine Department, BTT Medical Institute, Aventura, FL, United States
- BTT Engineering Department, BTT Medical Institute, Aventura, FL, United States
| | - Alberto F Chocron
- Medicine Department, BTT Medical Institute, Aventura, FL, United States
- Research Service, Miami Veteran Administration Medical Center, Miami, FL, United States
| | - David M Smadja
- Department of Hematology, AP-HP, Georges Pompidou European Hospital, Paris, France
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
4
|
Çolak R, Gültürk İ, Dinç G, Akdağ G, Yıldırım S, Yılmaz M, Tural D. The relationship between body mass index and survival in patients with renal cell carcinoma treated with nivolumab. J Chemother 2025:1-7. [PMID: 39988757 DOI: 10.1080/1120009x.2025.2469371] [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: 10/25/2024] [Revised: 01/02/2025] [Accepted: 02/07/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Immunotherapies have increased the therapeutic options for patients with metastatic renal cell carcinoma (mRCC), but the absence of prognostic indicators remains unresolved. We assessed the potential association of BMI with the overall survival (OS) of patients treated with nivolumab. METHODS We performed a retrospective study of 126 patients with histologically confirmed mRCC who began systemic ICI therapy between January 2016 and April 2022 were included. BMI at the time of treatment start was calculated. Then patients were divided into two groups: high BMI (≥25) and low BMI (<25). Therapeutic responses were determined according to RECIST v1.1. OS was defined as the time from starting ICI treatment until death or last follow-up at the data cutoff. RESULTS The cohort was male 74.6%, with a median age of 62. The median follow-up time was 18.6 months. The patients were classified as low BMI (<25) and high BMI (≥25). The OS was 40.6 months (95% CI: 34.2-47.0) for patients with high BMI vs. 9.4 months (95% CI: 7.0-11.7) for patients with low BMI, and a significant association was found between BMI and OS (p < 0.001). CONCLUSIONS BMI was an independent prognostic factor in the patients with mRCC treated with nivolumab. Prospective and multicenter research is needed to confirm our findings.
Collapse
Affiliation(s)
- Rumeysa Çolak
- Department of Medical Oncology, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - İlkay Gültürk
- Department of Medical Oncology, Saglik Bilimleri University, Istanbul Training and Research Hospital, Istanbul, Turkey
| | - Gülhan Dinç
- Department of Medical Oncology, Saglik Bilimleri University Professor Dr. Cemil Taşcıoğlu City Hospital, Istanbul, Turkey
| | - Goncagül Akdağ
- Department of Medical Oncology, Kartal Dr. Lütfi Kırdar City Hospital, Istanbul, Turkey
| | - Sedat Yıldırım
- Department of Medical Oncology, Kartal Dr. Lütfi Kırdar City Hospital, Istanbul, Turkey
| | - Mesut Yılmaz
- Department of Medical Oncology, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Deniz Tural
- Department of Medical Oncology, Koc University Hospital, Istanbul, Turkey
| |
Collapse
|
5
|
Fang Q, Qian Y, Xie Z, Zhao H, Zheng Y, Li D. Predictors of severity and onset timing of immune-related adverse events in cancer patients receiving immune checkpoint inhibitors: a retrospective analysis. Front Immunol 2025; 16:1508512. [PMID: 40040713 PMCID: PMC11876122 DOI: 10.3389/fimmu.2025.1508512] [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: 10/09/2024] [Accepted: 02/03/2025] [Indexed: 03/06/2025] Open
Abstract
Objective To identify predictors of all-grade, grade ≥ 3, and onset time of immune-related adverse events (irAEs) in cancer patients undergoing immune checkpoint inhibitors (ICIs) therapy. Methods This retrospective analysis included cancer patients treated with ICIs at Chongqing Medical University Second Affiliated Hospital from 2018 to 2024. Logistic regression and Cox regression analyses were used to identify predictors of all-grade and grade ≥ 3 irAEs and the time of irAE onset. Results Among the 3,795 patients analyzed, 1,101 (29.0%) developed all-grade irAEs, and 175 (4.6%) experienced grade ≥ 3 irAEs. Multivariate logistic regression revealed that female (OR = 1.37, p < 0.001), combination therapy (OR = 1.87, p < 0.001), pre-existing autoimmune diseases (AIDs) (OR = 5.15, p < 0.001), pre-existing cirrhosis (OR = 1.34, p = 0.001), antibiotic use during ICIs treatment (OR = 1.51, p < 0.001), and a higher baseline prognostic nutritional index (PNI) (OR = 1.23, p = 0.01) were significant predictors for the development of all-grade irAEs. The predictors for grade ≥ 3 irAEs included age ≥ 60 (OR = 1.49, p = 0.023) and pre-existing AIDs (OR = 2.09, p = 0.005), For the onset time, predictors included female (HR = 1.26, p = 0.001), combination therapy (HR = 1.80, p < 0.001), pre-existing AIDs (HR = 2.25, p < 0.001), and pre-existing infection (HR = 1.20, p = 0.008). Conclusions Females, combination therapy, pre-existing AIDs and cirrhosis, antibiotics, and a higher baseline PNI are associated with a higher risk of developing all-grade irAEs. Those aged ≥ 60 and with pre-existing AIDs face a higher risk of severe irAEs. Females, undergoing combination therapy, with pre-existing AIDs and infection generally experience a shorter time to irAEs onset. Multicentric prospective studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Qimei Fang
- Department of Pharmacy, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yan Qian
- Department of Pharmacy, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhaolu Xie
- Department of Pharmacy, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hongqiong Zhao
- Department of Pharmacy, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yang Zheng
- Department of Pharmacy, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Di Li
- Department of Pharmacy, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Szwed M, Jost T, Majka E, Gharibkandi NA, Majkowska-Pilip A, Frey B, Bilewicz A, Fietkau R, Gaipl U, Marczak A, Lubgan D. Pt-Au Nanoparticles in Combination with Near-Infrared-Based Hyperthermia Increase the Temperature and Impact on the Viability and Immune Phenotype of Human Hepatocellular Carcinoma Cells. Int J Mol Sci 2025; 26:1574. [PMID: 40004038 PMCID: PMC11855494 DOI: 10.3390/ijms26041574] [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: 12/31/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Near-infrared light (NIR)-responsive metal-based nanoparticles (NPs) could be used for tumour therapy. We examined how platinum (Pt), gold (Au), and core-shell Pt-Au NPs affect the viability of human hepatocellular carcinoma (HCC) cell lines (Hep3B, HepG2, and Huh7D-12) alone and in combination with NIR exposure. In addition, the expression of immune checkpoint molecules (ICMs) on the tumour cells was analysed. We revealed that the cytotoxicity and programmed cell death induction of Au and Pt-Au NPs toward HCC cells could be enhanced by NIR with 960 nm in a different way. Pt-Au NPs were the only particles that resulted in an additional temperature increase of up to 2 °C after NIR. Regarding the tumour cell immune phenotype, not all of the cells experienced changes in immune phenotype. NIR itself was the trigger of the alterations, while the NPs did not significantly affect the expression of most of the examined ICMs, such as PD-L1, PD-L1, HVEM, CD70, ICOS-L, Ox40-L, and TNFRSF9. The combination of Pt-Au NPs with NIR resulted in the most prominent increase of ICMs in HepG2 cells. We conclude that the thermotherapeutic effect of Pt-Au NP application and NIR could be beneficial in multimodal therapy settings in liver cancer for selected patients.
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Tina Jost
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (T.J.); (B.F.); (U.G.); (D.L.)
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Emilia Majka
- Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (E.M.); (N.A.G.); (A.M.-P.); (A.B.)
| | - Nasrin Abbasi Gharibkandi
- Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (E.M.); (N.A.G.); (A.M.-P.); (A.B.)
| | - Agnieszka Majkowska-Pilip
- Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (E.M.); (N.A.G.); (A.M.-P.); (A.B.)
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (T.J.); (B.F.); (U.G.); (D.L.)
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Aleksander Bilewicz
- Institute of Nuclear Chemistry and Technology, 03-195 Warsaw, Poland; (E.M.); (N.A.G.); (A.M.-P.); (A.B.)
| | - Rainer Fietkau
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Udo Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (T.J.); (B.F.); (U.G.); (D.L.)
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Agnieszka Marczak
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Dorota Lubgan
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany; (T.J.); (B.F.); (U.G.); (D.L.)
- Comprehensive Cancer Center Erlangen-EMN, D-91054 Erlangen, Germany;
- Department of Radiation Oncology, Universitatsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie, D-91054 Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| |
Collapse
|
7
|
Hiep Tran T, Thu Phuong Tran T. Current status of nanoparticle-mediated immunogenic cell death in cancer immunotherapy. Int Immunopharmacol 2024; 142:113085. [PMID: 39276455 DOI: 10.1016/j.intimp.2024.113085] [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: 06/24/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024]
Abstract
Immunogenic cell death (ICD) encompasses various forms of cell death modalities, including apoptosis, necroptosis, ferroptosis, and pyroptosis. It arises from a harmonious interplay of adjuvant (damage-associated molecular patterns-DAMPs and chemokines/cytokines) and antigenicity (tumor-associated antigens-TAA) to induce immune-reaction toward cancer cells. Inducing ICD stands out as a promising approach in cancer immunotherapy, capable of directly eliminating cancer cells and of eliciting enduring antitumor immune responses. Conventional tumor therapies like radiation therapy, photodynamic therapy, and chemotherapy can also induce ICD which could amplify their activities. The development of effective ICD inducers like nano-systems is crucial for ensuring safe and efficacious immunotherapy. Nanoparticles hold considerable promise in cancer therapy, offering enhanced therapeutic outcomes and mitigated side effects. They could be the capacity to adjust systemic biodistribution, augment the accumulation of therapeutic agents at the intended site and protect active agents from the complexity of human biofluid. This review aims to outline the role of nanoparticles in triggering ICD for cancer immunotherapy that potentially pave the way for cancer treatment.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam
| | - Thi Thu Phuong Tran
- Department of Life Sciences, University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam.
| |
Collapse
|
8
|
Tzang CC, Lee YW, Lin WC, Lin LH, Kang YF, Lin TY, Wu WT, Chang KV. Evaluation of immune checkpoint inhibitors for colorectal cancer: A network meta‑analysis. Oncol Lett 2024; 28:569. [PMID: 39390977 PMCID: PMC11465421 DOI: 10.3892/ol.2024.14702] [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: 07/10/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Colorectal cancer (CRC) is challenging to treat due to its high metastatic rate. Recent strategies have focused on combining immune checkpoint inhibitors (ICIs) with other treatments. The aim of the present study was to conduct a network meta-analysis of randomized controlled trials (RCTs) to assess the efficacy and adverse effects of different ICI treatments for CRC. A literature search for RCTs was conducted using PubMed, the Cochrane Library, Embase, ClinicalTrials.gov and Web of Science databases, covering the period from the inception of each database until April 2024. A total of 12 RCTs involving 2,050 participants were selected for inclusion in the analysis. The network meta-analysis employed the MetaInsight tool to assess multiple endpoints. The criteria for study selection were based on the Population, Intervention, Comparison, Outcome and Studies framework as follows: i) Population, patients with CRC; ii) intervention, studies using ICI to treat CRC; iii) comparison, active comparators, including placebo; iv) outcome, overall survival, progression-free survival, objective response rate and adverse events; and v) study design, RCTs. The results of the analysis revealed that programmed cell death-ligand 1 (PD-L1) inhibitors significantly improved overall survival time [mean difference (MD), 2.28 months; 95% confidence interval (CI), 0.44 to 4.11], while programmed cell death protein 1 (PD-1) inhibitors exhibited a superior progression-free survival time (MD, 4.79 months; 95% CI, 3.18 to 6.40) compared with active comparators. However, none of the ICI treatments had significant differences in odds ratios for the objective response rate and adverse events compared with active comparators. These findings indicate that treatment with PD-L1 and PD-1 inhibitors improved the overall survival time and delayed disease progression in patients with CRC. These findings offer valuable insights for future research aimed at improving CRC patient outcomes.
Collapse
Affiliation(s)
- Chih-Chen Tzang
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan, R.O.C
| | - Yen-Wei Lee
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan, R.O.C
| | - Wei-Chen Lin
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan, R.O.C
| | - Long-Huei Lin
- School of Physical Therapy and Graduate Institute of Rehabilitation Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, R.O.C
| | - Yuan-Fu Kang
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan, R.O.C
| | - Ting-Yu Lin
- Department of Physical Medicine and Rehabilitation, Lo-Hsu Medical Foundation, Inc., Lotung Poh-Ai Hospital, Yilan 265, Taiwan, R.O.C
| | - Wei-Ting Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan, R.O.C
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei 108, Taiwan, R.O.C
| | - Ke-Vin Chang
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan, R.O.C
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei 108, Taiwan, R.O.C
- Center for Regional Anesthesia and Pain Medicine, Wang-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan, R.O.C
| |
Collapse
|
9
|
Ye M, Yin D, Wu Y, Miao H, Wu Z, Liu P. Infrared radiation for cancer hyperthermia: the light to brighten up oncology. Expert Rev Anticancer Ther 2024; 24:1147-1160. [PMID: 39390965 DOI: 10.1080/14737140.2024.2416063] [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: 07/20/2024] [Revised: 09/09/2024] [Accepted: 10/09/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Cancer constitutes the greatest public health threat to humans, as its incidence and mortality rates continue to increase worldwide. With the development of medical physics, more practitioners focus on the direct and indirect anti-tumor effects of physical factors. Infrared radiation (INR) is currently the most rapidly developing physical therapy method for tumors and has become a favored target for many oncologists and researchers owing to its advantages of high efficiency, low toxicity, and strong feasibility. AREAS COVERED This work provides a comprehensive collection of the latest information on INR anti-tumor research, drawing from public medical databases (PubMed, Web of Science, Embase, and Clinical Trials) from the last 10 years (2014 to 2024), and encompassing both basic and clinical research in oncology and physics. This article reviews the application of INR in tumor hyperthermia, summarizes and analyzes the practical value of INR for tumor treatment, and discusses future development trends to provide valuable assistance for the subsequent development of oncology. EXPERT OPINION Currently, INR has continuously accumulated excellent data in the field of tumor hyperthermia, bringing practical survival benefits to patients with cancer, and playing an important role in basic and clinical cancer research.
Collapse
Affiliation(s)
- Mengna Ye
- Department of Internal Medicine, Liangzhu Sub-District Community Health Service Center, Hangzhou, China
| | - Dashan Yin
- Department of Radiation Oncology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Yufei Wu
- ACS (International) School of Singapore, Singapore, Singapore
| | - Hua Miao
- Department of Internal Medicine, Liangzhu Sub-District Community Health Service Center, Hangzhou, China
| | - Zhibing Wu
- Department of Oncology, Zhejiang Hospital, Hangzhou, China
- Department of Internal Medicine, Liangzhu Sub-District Community Health Service Center, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Pengyuan Liu
- Department of Oncology, Zhejiang Hospital, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Righini MF, Durham A, Tsoutsou PG. Hyperthermia and radiotherapy: physiological basis for a synergistic effect. Front Oncol 2024; 14:1428065. [PMID: 39165690 PMCID: PMC11333208 DOI: 10.3389/fonc.2024.1428065] [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: 05/05/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024] Open
Abstract
In cancer treatment, mild hyperthermia (HT) represents an old, but recently revived opportunity to increase the efficacy of radiotherapy (RT) without increasing side effects, thereby widening the therapeutic window. HT disrupts cellular homeostasis by acting on multiple targets, and its combination with RT produces synergistic antitumoral effects on specific pathophysiological mechanisms, associated to DNA damage and repair, hypoxia, stemness and immunostimulation. HT is furthermore associated to direct tumor cell kill, particularly in higher temperature levels. A phenomenon of temporary resistance to heat, known as thermotolerance, follows each HT session. Cancer treatment requires innovative concepts and combinations to be tested but, for a meaningful development of clinical trials, the understanding of the underlying mechanisms of the tested modalities is essential. In this mini-review, we aimed to describe the synergistic effects of the combination of HT with RT as well as the phenomena of thermal shock and thermotolerance, in order to stimulate clinicians in new, clinically relevant concepts and combinations, which become particularly relevant in the era of technological advents in both modalities but also cancer immunotherapy.
Collapse
Affiliation(s)
| | - André Durham
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Radiation Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Pelagia G. Tsoutsou
- Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Radiation Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| |
Collapse
|
11
|
Donoso-González O, Riveros AL, Marco JF, Venegas-Yazigi D, Paredes-García V, Olguín CF, Mayorga-Lobos C, Lobos-González L, Franco-Campos F, Wang J, Kogan MJ, Bollo S, Yañez C, Báez DF. Iron-Reduced Graphene Oxide Core-Shell Micromotors Designed for Magnetic Guidance and Photothermal Therapy under Second Near-Infrared Light. Pharmaceutics 2024; 16:856. [PMID: 39065553 PMCID: PMC11279713 DOI: 10.3390/pharmaceutics16070856] [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: 05/14/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Core-shell micro/nanomotors have garnered significant interest in biomedicine owing to their versatile task-performing capabilities. However, their effectiveness for photothermal therapy (PTT) still faces challenges because of their poor tumor accumulation, lower light-to-heat conversion, and due to the limited penetration of near-infrared (NIR) light. In this study, we present a novel core-shell micromotor that combines magnetic and photothermal properties. It is synthesized via the template-assisted electrodeposition of iron (Fe) and reduced graphene oxide (rGO) on a microtubular pore-shaped membrane. The resulting Fe-rGO micromotor consists of a core of oval-shaped zero-valent iron nanoparticles with large magnetization. At the same time, the outer layer has a uniform reduced graphene oxide (rGO) topography. Combined, these Fe-rGO core-shell micromotors respond to magnetic forces and near-infrared (NIR) light (1064 nm), achieving a remarkable photothermal conversion efficiency of 78% at a concentration of 434 µg mL-1. They can also carry doxorubicin (DOX) and rapidly release it upon NIR irradiation. Additionally, preliminary results regarding the biocompatibility of these micromotors through in vitro tests on a 3D breast cancer model demonstrate low cytotoxicity and strong accumulation. These promising results suggest that such Fe-rGO core-shell micromotors could hold great potential for combined photothermal therapy.
Collapse
Affiliation(s)
- Orlando Donoso-González
- Departamento Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile; (O.D.-G.); (A.L.R.); (M.J.K.); (S.B.)
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile;
| | - Ana L. Riveros
- Departamento Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile; (O.D.-G.); (A.L.R.); (M.J.K.); (S.B.)
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile;
| | - José F. Marco
- Instituto de Química Física Blas Cabrera, Consejo Superior de Investigaciones Científicas (CSIC), Serrano 119, 28006 Madrid, Spain;
| | - Diego Venegas-Yazigi
- Departamento de Química de los Materiales, Facultad de Química y Biología, Universidad de Santiago de Chile, Libertador Bernardo O’Higgins #3363, Estación Central, Santiago 9170022, Chile;
- Centro para el Desarrollo de La Nanociencia y la Nanotecnología (CEDENNA), Universidad de Santiago de Chile, Libertador Bernardo O’Higgins #3363, Estación Central, Santiago 9170022, Chile;
| | - Verónica Paredes-García
- Centro para el Desarrollo de La Nanociencia y la Nanotecnología (CEDENNA), Universidad de Santiago de Chile, Libertador Bernardo O’Higgins #3363, Estación Central, Santiago 9170022, Chile;
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andrés Bello, República 275, Santiago, Santiago 8370146, Chile
| | - Camila F. Olguín
- Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile;
| | - Cristina Mayorga-Lobos
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile;
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380492, Chile;
| | - Lorena Lobos-González
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago 8380492, Chile;
- Center for Regenerative Medicine, Institute for Sciences and Innovation in Medicine, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Felipe Franco-Campos
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, 46100 Valencia, Spain;
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA 92093, USA;
| | - Marcelo J. Kogan
- Departamento Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile; (O.D.-G.); (A.L.R.); (M.J.K.); (S.B.)
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile;
| | - Soledad Bollo
- Departamento Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile; (O.D.-G.); (A.L.R.); (M.J.K.); (S.B.)
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile;
- Centro de Investigación de Procesos Redox, CIPRex, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile
| | - Claudia Yañez
- Centro de Investigación de Procesos Redox, CIPRex, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile
- Departamento de Química Orgánica y Fisicoquímica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile
| | - Daniela F. Báez
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone #1007, Independencia, Santiago 8380492, Chile;
- Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile;
| |
Collapse
|
12
|
Wang L, Chen J, Ma C, Zhang C. Targeted nanotherapy platform mediated tumor-infiltrating CD8 + T cell immune function effects for collaborative anti-tumor photothermal immunotherapy for cervical cancer. NANOSCALE ADVANCES 2024; 6:3052-3063. [PMID: 38868823 PMCID: PMC11166113 DOI: 10.1039/d3na01132a] [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: 12/19/2023] [Accepted: 04/10/2024] [Indexed: 06/14/2024]
Abstract
Photothermal immunotherapy is an innovative approach to cancer treatment. It combines immunomodulators and photothermal agents, both targeted to the tumor site. This therapy harnesses the heat generated by photothermal conversion to damage tumor cells while simultaneously releasing tumor-associated antigens. This process enhances the anti-tumor immune response of tumor-infiltrating lymphocytes (TILs) within the tumor microenvironment (TME). Photothermal immunotherapy is gaining prominence as a new method for cancer treatment. It is a current focal point in research due to its targeted efficacy, minimal systemic side effects, and reduced risk of treatment resistance. This study employed a thin-film dispersion method to fabricate liposomes (LIPO) as composite drug carriers. Indocyanine green (ICG) for clinical use was utilized as a photothermal agent (PTA), and folate (FA) was employed as a targeting agent for the nano-composite material. We encapsulated the immunoadjuvant CpG ODN within the FA@LIPO@ICG nano-system, resulting in the formation of targeted nanoparticles (NPs) for photothermal immunotherapy (FA@LIPO@ICG@CpG), and assessed the drug encapsulation rate. FA@LIPO@ICG@CpG NPs demonstrated excellent water solubility with an average size ranging from 100 to 200 nm. Furthermore, we investigated the photothermal properties of FA@LIPO@ICG@CpG NPs. Under 808 nm laser irradiation, the photothermal conversion efficiency of FA@LIPO@ICG@CpG NPs reached 39.05%. Subsequently, under 808 nm laser excitation, we conducted an analysis of lymphocyte subpopulations and their functional changes in U14 tumor-bearing mice by using flow cytometry. This treatment approach demonstrated remarkable anti-tumor efficacy. Consequently, FA@LIPO@ICG@CpG NPs hold substantial promise as a novel and promising strategy in cancer therapy.
Collapse
Affiliation(s)
- Lei Wang
- Department of Gynecology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia Urumqi 830054 Xinjiang China
| | - Jianhuan Chen
- Department of Gynecology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia Urumqi 830054 Xinjiang China
| | - Cailing Ma
- Department of Gynecology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia Urumqi 830054 Xinjiang China
| | - Chuanshan Zhang
- Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia Urumqi 830054 Xinjiang China
| |
Collapse
|
13
|
Uzdrowska K, Knap N, Gulczynski J, Kuban-Jankowska A, Struck-Lewicka W, Markuszewski MJ, Bączek T, Izycka-Swieszewska E, Gorska-Ponikowska M. Chasing Graphene-Based Anticancer Drugs: Where are We Now on the Biomedical Graphene Roadmap? Int J Nanomedicine 2024; 19:3973-3989. [PMID: 38711615 PMCID: PMC11073537 DOI: 10.2147/ijn.s447397] [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: 10/31/2023] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
Graphene and graphene-based materials have attracted growing interest for potential applications in medicine because of their good biocompatibility, cargo capability and possible surface functionalizations. In parallel, prototypic graphene-based devices have been developed to diagnose, imaging and track tumor growth in cancer patients. There is a growing number of reports on the use of graphene and its functionalized derivatives in the design of innovative drugs delivery systems, photothermal and photodynamic cancer therapy, and as a platform to combine multiple therapies. The aim of this review is to introduce the latest scientific achievements in the field of innovative composite graphene materials as potentially applied in cancer therapy. The "Technology and Innovation Roadmap" published in the Graphene Flagship indicates, that the first anti-cancer drugs using graphene and graphene-derived materials will have appeared on the market by 2030. However, it is necessary to broaden understanding of graphene-based material interactions with cellular metabolism and signaling at the functional level, as well as toxicity. The main aspects of further research should elucidate how treatment methods (e.g., photothermal therapy, photodynamic therapy, combination therapy) and the physicochemical properties of graphene materials influence their ability to modulate autophagy and kill cancer cells. Interestingly, recent scientific reports also prove that graphene nanocomposites modulate cancer cell death by inducing precise autophagy dysfunctions caused by lysosome damage. It turns out as well that developing photothermal oncological treatments, it should be taken into account that near-infrared-II radiation (1000-1500 nm) is a better option than NIR-I (750-1000 nm) because it can penetrate deeper into tissues due to less scattering at longer wavelengths radiation.
Collapse
Affiliation(s)
- Katarzyna Uzdrowska
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk, 80-211, Poland
| | - Narcyz Knap
- Department of Medical Chemistry, Medical University of Gdansk, Gdansk, 80-211, Poland
| | - Jacek Gulczynski
- Faculty of Health Sciences with the Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, 80-211, Poland
| | | | | | | | - Tomasz Bączek
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, 80-416, Poland
| | - Ewa Izycka-Swieszewska
- Faculty of Health Sciences with the Institute of Maritime and Tropical Medicine, Medical University of Gdansk, Gdansk, 80-211, Poland
| | | |
Collapse
|
14
|
Liu J, Zhang G, Li X, Zheng C, Kan X. Enhancing the therapeutic impact of sublethal radiofrequency hyperthermia in malignant solid tumor treatment. Heliyon 2024; 10:e29866. [PMID: 38681568 PMCID: PMC11053292 DOI: 10.1016/j.heliyon.2024.e29866] [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: 02/13/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024] Open
Abstract
Radiofrequency ablation (RFA) is an effective alternative to surgery for managing some malignant solid tumors. However, for medium-to-large tumors (>3 cm), tumors adjacent to large blood vessels, and certain irregular tumors, sublethal radiofrequency hyperthermia (RFH) often produces a margin of ablated tumor owing to the "heat-sink" effect. This effect typically leaves behind viable residual tumors at the margin. Several studies have reported that a sublethal RFH can significantly enhance the efficacy of chemotherapy, radiotherapy, immunotherapy, and gene therapy for malignant solid tumors. The possible mechanisms by which RFH enhances these therapies include heat-induced tissue fracturing, increased permeability of the cytoplasmic membrane, exaggerated cellular metabolism, blockade of the repair pathways of radiation-damaged tumor cells, and activation of the heat shock protein pathways. Therefore, RFA in combination with chemotherapy, radiotherapy, immunotherapy, or gene therapy may help reduce the rates of residual and recurrent tumors after RFA of malignant solid tumors.
Collapse
Affiliation(s)
- Jiayun Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Guilin Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xinyi Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
15
|
Croitoru AM, Ficai D, Ficai A. Novel Photothermal Graphene-Based Hydrogels in Biomedical Applications. Polymers (Basel) 2024; 16:1098. [PMID: 38675017 PMCID: PMC11053615 DOI: 10.3390/polym16081098] [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: 03/11/2024] [Revised: 03/27/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
In the last decade, photothermal therapy (PTT) has attracted tremendous attention because it is non-invasive, shows high efficiency and antibacterial activity, and minimizes drug side effects. Previous studies demonstrated that PTT can effectively inhibit the growth of bacteria and promotes cell proliferation, accelerating wound healing and tissue regeneration. Among different NIR-responsive biomaterials, graphene-based hydrogels with photothermal properties are considered as the best candidates for biomedical applications, due to their excellent properties. This review summarizes the current advances in the development of innovative graphene-based hydrogels for PTT-based biomedical applications. Also, the information about photothermal properties and the potential applications of graphene-based hydrogels in biomedical therapies are provided. These findings provide a great potential for supporting their applications in photothermal biomedicine.
Collapse
Affiliation(s)
- Alexa-Maria Croitoru
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, Spl. Independentei 91-95, 0500957 Bucharest, Romania;
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, National University for Science and Technology Politehnica Bucharest, Gh. Polizu St. 1-7, 011061 Bucharest, Romania;
- National Centre for Food Safety, National University for Science and Technology Politehnica Bucharest, Spl. Independentei 313, 060042 Bucharest, Romania
| | - Denisa Ficai
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, National University for Science and Technology Politehnica Bucharest, Gh. Polizu St. 1-7, 011061 Bucharest, Romania;
- National Centre for Food Safety, National University for Science and Technology Politehnica Bucharest, Spl. Independentei 313, 060042 Bucharest, Romania
| | - Anton Ficai
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Chemical Engineering and Biotechnologies, National University for Science and Technology Politehnica Bucharest, Gh. Polizu St. 1-7, 011061 Bucharest, Romania;
- National Centre for Micro- and Nanomaterials, National University for Science and Technology Politehnica Bucharest, Spl. Independentei 313, 060042 Bucharest, Romania
- Academy of Romanian Scientists, 3 Ilfov Street, 050045 Bucharest, Romania
| |
Collapse
|
16
|
Liu P, Wei Z, Ye X. Immunostimulatory effects of thermal ablation: Challenges and future prospects. J Cancer Res Ther 2024; 20:531-539. [PMID: 38687922 DOI: 10.4103/jcrt.jcrt_2484_23] [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: 11/18/2023] [Accepted: 12/13/2023] [Indexed: 05/02/2024]
Abstract
ABSTRACT This literature explores the immunostimulatory effects of thermal ablation in the tumor microenvironment, elucidating the mechanisms such as immunogenic cell death, tumor-specific antigens, and damage-associated molecular patterns. Furthermore, it outlines critical issues associated with thermal ablation-induced immunostimulatory challenges and offers insights into future research avenues and potential therapeutic strategies.
Collapse
Affiliation(s)
- Peng Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, No. 16766 Jingshi Road, Jinan, Shandong Province, China
| | | | | |
Collapse
|
17
|
Lukácsi S, Munkácsy G, Győrffy B. Harnessing Hyperthermia: Molecular, Cellular, and Immunological Insights for Enhanced Anticancer Therapies. Integr Cancer Ther 2024; 23:15347354241242094. [PMID: 38818970 PMCID: PMC11143831 DOI: 10.1177/15347354241242094] [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: 12/28/2023] [Revised: 02/25/2024] [Accepted: 03/11/2024] [Indexed: 06/01/2024] Open
Abstract
Hyperthermia, the raising of tumor temperature (≥39°C), holds great promise as an adjuvant treatment for cancer therapy. This review focuses on 2 key aspects of hyperthermia: its molecular and cellular effects and its impact on the immune system. Hyperthermia has profound effects on critical biological processes. Increased temperatures inhibit DNA repair enzymes, making cancer cells more sensitive to chemotherapy and radiation. Elevated temperatures also induce cell cycle arrest and trigger apoptotic pathways. Furthermore, hyperthermia modifies the expression of heat shock proteins, which play vital roles in cancer therapy, including enhancing immune responses. Hyperthermic treatments also have a significant impact on the body's immune response against tumors, potentially improving the efficacy of immune checkpoint inhibitors. Mild systemic hyperthermia (39°C-41°C) mimics fever, activating immune cells and raising metabolic rates. Intense heat above 50°C can release tumor antigens, enhancing immune reactions. Using photothermal nanoparticles for targeted heating and drug delivery can also modulate the immune response. Hyperthermia emerges as a cost-effective and well-tolerated adjuvant therapy when integrated with immunotherapy. This comprehensive review serves as a valuable resource for the selection of patient-specific treatments and the guidance of future experimental studies.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Gyöngyi Munkácsy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
| | - Balázs Győrffy
- HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Semmelweis University, Budapest, Hungary
- University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| |
Collapse
|
18
|
Yu H, Zhang X, Li J, Wang K, Yin C, Li X, Li L, Shao G, Jin S. Design, Synthesis and Evaluation of a Novel Teoptinib Derivative as an Effective Anti-hepatocellular Carcinoma Agent. Curr Pharm Des 2024; 30:2167-2178. [PMID: 38919077 DOI: 10.2174/0113816128314500240621071306] [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: 03/14/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND & PURPOSE Hepatocellular Carcinoma (HCC) is a type of liver cancer known for its poor prognosis and high mortality. Teoptinib is a highly selective MET inhibitor that has been used in the treatment of liver cancer. Although good progress has been made in clinical treatment, further improvement is still needed. In this study, a series of novel Teoptinib derivatives were synthesized and evaluated as anti-cancer agents for the treatment of liver cancer, and an oral nanodrug delivery system was also explored. METHODS A series of novel Teoptinib derivatives were synthesized, and an oral nanodrug delivery system was also explored. HPLC, high-resolution mass spectrometer and NMR were used to determine the structure and molecular formula of the synthesized compounds. Zeta potential assay was used to access the particle size distribution and zeta potential of the nanoparticles. MTT assay, cell colony formation assay, cell apoptosis inhibition assay, cell scratch assay, and the MHCC-97H xenograft model of nude mice assay were used to evaluate the in vitro and in vivo anti-tumor activity of the synthesized compounds. RESULTS Compound (R)-10 showed the best antitumor activity with 0.010 μM of the IC50 value against MHCC-97H, a human liver cancer cell line with high c-Met expression. The MHCC-97H xenograft model of nude mice assay showed that nano-prodrug of compound (R)-10 exhibited good in vivo activity with 87.67% of the TGI at the dosage of 8 mg/kg. CONCLUSION We designed and synthesized a series of c-Met inhibitors containing different side chains and chiral centers as anti-liver cancer agents. Among them, compound (R)-10 shows a promising effect as a lead molecule for further study in the treatment of liver cancer. The successful incorporation of (R)-10 into a novel oral nanodrug delivery system highlights the importance of effective drug delivery systems for enhanced therapeutic efficacy.
Collapse
MESH Headings
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Animals
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Drug Design
- Mice
- Mice, Nude
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Drug Screening Assays, Antitumor
- Structure-Activity Relationship
- Molecular Structure
- Dose-Response Relationship, Drug
- Proto-Oncogene Proteins c-met/antagonists & inhibitors
- Proto-Oncogene Proteins c-met/metabolism
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/metabolism
- Mice, Inbred BALB C
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Cell Line, Tumor
- Nanoparticles/chemistry
Collapse
Affiliation(s)
- Huijuan Yu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510275, China
| | - Xiaodong Zhang
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China
| | - Jiayu Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510275, China
| | - Kaimei Wang
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Changjun Yin
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xinshu Li
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China
| | - Lianyun Li
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China
| | - Guang Shao
- School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China
| | - Shaowen Jin
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| |
Collapse
|
19
|
Lei L, Liu P, Jing W, Wu Z. Advancements in modifying the efficacy of immunotherapies through the thermal effects of nanomaterials. NANO TRANSMED 2023; 2:100022. [DOI: 10.1016/j.ntm.2023.100022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
|
20
|
Báez DF. Graphene-Based Nanomaterials for Photothermal Therapy in Cancer Treatment. Pharmaceutics 2023; 15:2286. [PMID: 37765255 PMCID: PMC10535159 DOI: 10.3390/pharmaceutics15092286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/17/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Graphene-based nanomaterials (GBNMs), specifically graphene oxide (GO) and reduced graphene oxide (rGO), have shown great potential in cancer therapy owing to their physicochemical properties. As GO and rGO strongly absorb light in the near-infrared (NIR) region, they are useful in photothermal therapy (PTT) for cancer treatment. However, despite the structural similarities of GO and rGO, they exhibit different influences on anticancer treatment due to their different photothermal capacities. In this review, various characterization techniques used to compare the structural features of GO and rGO are first outlined. Then, a comprehensive summary and discussion of the applicability of GBNMs in the context of PTT for diverse cancer types are presented. This discussion includes the integration of PTT with secondary therapeutic strategies, with a particular focus on the photothermal capacity achieved through near-infrared irradiation parameters and the modifications implemented. Furthermore, a dedicated section is devoted to studies on hybrid magnetic-GBNMs. Finally, the challenges and prospects associated with the utilization of GBNM in PTT, with a primary emphasis on the potential for clinical translation, are addressed.
Collapse
Affiliation(s)
- Daniela F. Báez
- Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile;
- Instituto de Investigación Interdisciplinaria, Vicerrectoría Académica, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
21
|
Obrador E, Montoro A. Ionizing Radiation, Antioxidant Response and Oxidative Damage: Radiomodulators. Antioxidants (Basel) 2023; 12:1219. [PMID: 37371949 DOI: 10.3390/antiox12061219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Ionizing radiation (IR) is the energy released by atoms in the form of electromagnetic waves (e [...].
Collapse
Affiliation(s)
- Elena Obrador
- Elena Obrador Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
| | - Alegría Montoro
- Alegría Montoro, Radiation Protection Service, University and Polytechnic Hospital La Fe, 46021 Valencia, Spain
| |
Collapse
|
22
|
Kaur I, Tieu T, Deepagan VG, Ali MA, Alsunaydih F, Rudd D, Moghaddam MA, Bourgeois L, Adams TE, Thurecht KJ, Yuce M, Cifuentes-Rius A, Voelcker NH. Combination of Chemotherapy and Mild Hyperthermia Using Targeted Nanoparticles: A Potential Treatment Modality for Breast Cancer. Pharmaceutics 2023; 15:pharmaceutics15051389. [PMID: 37242631 DOI: 10.3390/pharmaceutics15051389] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/17/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Despite the clinical benefits that chemotherapeutics has had on the treatment of breast cancer, drug resistance remains one of the main obstacles to curative cancer therapy. Nanomedicines allow therapeutics to be more targeted and effective, resulting in enhanced treatment success, reduced side effects, and the possibility of minimising drug resistance by the co-delivery of therapeutic agents. Porous silicon nanoparticles (pSiNPs) have been established as efficient vectors for drug delivery. Their high surface area makes them an ideal carrier for the administration of multiple therapeutics, providing the means to apply multiple attacks to the tumour. Moreover, immobilising targeting ligands on the pSiNP surface helps direct them selectively to cancer cells, thereby reducing harm to normal tissues. Here, we engineered breast cancer-targeted pSiNPs co-loaded with an anticancer drug and gold nanoclusters (AuNCs). AuNCs have the capacity to induce hyperthermia when exposed to a radiofrequency field. Using monolayer and 3D cell cultures, we demonstrate that the cell-killing efficacy of combined hyperthermia and chemotherapy via targeted pSiNPs is 1.5-fold higher than applying monotherapy and 3.5-fold higher compared to using a nontargeted system with combined therapeutics. The results not only demonstrate targeted pSiNPs as a successful nanocarrier for combination therapy but also confirm it as a versatile platform with the potential to be used for personalised medicine.
Collapse
Affiliation(s)
- Ishdeep Kaur
- Monash Institute of Pharmacy and Pharmaceutical Sciences, Monash University, 381, Royal Parade, Parkville, VIC 3052, Australia
| | - Terence Tieu
- Monash Institute of Pharmacy and Pharmaceutical Sciences, Monash University, 381, Royal Parade, Parkville, VIC 3052, Australia
| | - Veerasikku G Deepagan
- Monash Institute of Pharmacy and Pharmaceutical Sciences, Monash University, 381, Royal Parade, Parkville, VIC 3052, Australia
| | - Muhammad A Ali
- Department of Electrical and Computer Systems Engineering, Monash University, Clayton Campus, Clayton, VIC 3168, Australia
| | - Fahad Alsunaydih
- Department of Electrical and Computer Systems Engineering, Monash University, Clayton Campus, Clayton, VIC 3168, Australia
| | - David Rudd
- Monash Institute of Pharmacy and Pharmaceutical Sciences, Monash University, 381, Royal Parade, Parkville, VIC 3052, Australia
| | - Maliheh A Moghaddam
- Centre of Polymer Systems, Tomas Bata University, 5678, 760 01 Zlin, Czech Republic
| | - Laure Bourgeois
- Monash Centre for Electron Microscopy, Clayton Campus, Monash University, Clayton, VIC 3168, Australia
| | - Timothy E Adams
- Commonwealth Scientific and Industrial Research Organization (CSIRO), 343, Royal Parade, Parkville, VIC 3052, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology (AIBN), Corner College and Cooper Rds, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mehmet Yuce
- Department of Electrical and Computer Systems Engineering, Monash University, Clayton Campus, Clayton, VIC 3168, Australia
| | - Anna Cifuentes-Rius
- Monash Institute of Pharmacy and Pharmaceutical Sciences, Monash University, 381, Royal Parade, Parkville, VIC 3052, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmacy and Pharmaceutical Sciences, Monash University, 381, Royal Parade, Parkville, VIC 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC 3168, Australia
| |
Collapse
|
23
|
Mulens-Arias V, Nicolás-Boluda A, Carn F, Gazeau F. Cationic Polyethyleneimine (PEI)–Gold Nanocomposites Modulate Macrophage Activation and Reprogram Mouse Breast Triple-Negative MET-1 Tumor Immunological Microenvironment. Pharmaceutics 2022; 14:pharmaceutics14102234. [PMID: 36297669 PMCID: PMC9607133 DOI: 10.3390/pharmaceutics14102234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Nanomedicines based on inorganic nanoparticles have grown in the last decades due to the nanosystems’ versatility in the coating, tuneability, and physical and chemical properties. Nonetheless, concerns have been raised regarding the immunotropic profile of nanoparticles and how metallic nanoparticles affect the immune system. Cationic polymer nanoparticles are widely used for cell transfection and proved to exert an adjuvant immunomodulatory effect that improves the efficiency of conventional vaccines against infection or cancer. Likewise, gold nanoparticles (AuNPs) also exhibit diverse effects on immune response depending on size or coatings. Photothermal or photodynamic therapy, radiosensitization, and drug or gene delivery systems take advantage of the unique properties of AuNPs to deeply modify the tumoral ecosystem. However, the collective effects that AuNPs combined with cationic polymers might exert on their own in the tumor immunological microenvironment remain elusive. The purpose of this study was to analyze the triple-negative breast tumor immunological microenvironment upon intratumoral injection of polyethyleneimine (PEI)–AuNP nanocomposites (named AuPEI) and elucidate how it might affect future immunotherapeutic approaches based on this nanosystem. AuPEI nanocomposites were synthesized through a one-pot synthesis method with PEI as both a reducing and capping agent, resulting in fractal assemblies of about 10 nm AuNPs. AuPEI induced an inflammatory profile in vitro in the mouse macrophage-like cells RAW264.7 as determined by the secretion of TNF-α and CCL5 while the immunosuppressor IL-10 was not increased. However, in vivo in the mouse breast MET-1 tumor model, AuPEI nanocomposites shifted the immunological tumor microenvironment toward an M2 phenotype with an immunosuppressive profile as determined by the infiltration of PD-1-positive lymphocytes. This dichotomy in AuPEI nanocomposites in vitro and in vivo might be attributed to the highly complex tumor microenvironment and highlights the importance of testing the immunogenicity of nanomaterials in vitro and more importantly in vivo in relevant immunocompetent mouse tumor models to better elucidate any adverse or unexpected effect.
Collapse
Affiliation(s)
- Vladimir Mulens-Arias
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
- Integrative Biomedical Materials and Nanomedicine Lab, Department of Medicine and Life Sciences (MELIS), Pompeu Fabra University, PRBB, Carrer Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Alba Nicolás-Boluda
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
| | - Florent Carn
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
| | - Florence Gazeau
- Matière et Systèmes Complexes (MSC), Université Paris Cité, CNRS, 45 rue des Saints Pères, 75006 Paris, France
- Correspondence:
| |
Collapse
|