1
|
Fayyaz A, Haqqi A, Khan R, Irfan M, Khan K, Reiner Ž, Sharifi-Rad J, Calina D. Revolutionizing cancer treatment: the rise of personalized immunotherapies. Discov Oncol 2024; 15:756. [PMID: 39692978 DOI: 10.1007/s12672-024-01638-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Interest in biological therapy for cancer has surged due to its precise targeting of cancer cells and minimized impact on surrounding healthy tissues. This review discusses various biological cancer therapies, highlighting advanced alternatives over conventional chemotherapy alone. It explores DNA and RNA-based vaccines, T-cell modifications, adoptive cell transfer, CAR T cell therapy, angiogenesis inhibitors, and the combination of immunotherapy with chemotherapy, offering a holistic view of the potential in cancer treatment. Additionally, it discusses the role of nanotechnology in increasing the efficacy of cancer-targeting drugs, as well as cytokine and immunoconjugate therapies for bolstering immune system effectiveness against neoplastic cells. The potential of gene potential for precise targeting of cancer-linked genes and the application of oncolytic viruses against virus-associated cancers are also discussed. The review identifies significant advancements in the targeted treatment of cancer by biological methods. It acknowledges the challenges, including drug resistance and the need for high specificity in certain therapies, while also highlighting the effectiveness of cancer vaccines, modified T-cells, and oncolytic viruses. Biological therapies are a promising frontier in cancer treatment, offering the potential for more personalized and effective therapeutic strategies. Despite existing challenges, ongoing research and clinical trials are fundamental for overcoming current limitations and enhancing the efficacy of biological therapies in cancer care.
Collapse
Affiliation(s)
- Amna Fayyaz
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Aleena Haqqi
- School of Medical Laboratory Technology, Faculty of Allied Health Sciences, Minhaj University Lahore (MUL), Lahore, 54000, Pakistan
| | - Rashid Khan
- Department of Pharmacy, Punjab University College of Pharmacy University of Punjab Lahore, Lahore, 54000, Pakistan
| | - Muhammad Irfan
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Khushbukhat Khan
- Cancer Clinical Research Unit, Trials360, Lahore, 54000, Pakistan.
| | - Željko Reiner
- Department for Metabolic Diseases, University Hospital Center Zagreb, Zagreb, Croatia
- Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Centro de Estudios Tecnológicos, Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
2
|
Padzińska-Pruszyńska IB, Taciak B, Kiraga Ł, Smolarska A, Górczak M, Kucharzewska P, Kubiak M, Szeliga J, Matejuk A, Król M. Targeting Cancer: Microenvironment and Immunotherapy Innovations. Int J Mol Sci 2024; 25:13569. [PMID: 39769334 PMCID: PMC11679359 DOI: 10.3390/ijms252413569] [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: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
In 2024, the United States was projected to experience 2 million new cancer diagnoses and approximately 611,720 cancer-related deaths, reflecting a broader global trend in which cancer cases are anticipated to exceed 35 million by 2050. This increasing burden highlights ongoing challenges in cancer treatment despite significant advances that have reduced cancer mortality by 31% since 1991. Key obstacles include the disease's inherent heterogeneity and complexity, such as treatment resistance, cancer stem cells, and the multifaceted tumor microenvironment (TME). The TME-comprising various tumor and immune cells, blood vessels, and biochemical factors-plays a crucial role in tumor growth and resistance to therapies. Recent innovations in cancer treatment, particularly in the field of immuno-oncology, have leveraged insights into TME interactions. An emerging example is the FDA-approved therapy using tumor-infiltrating lymphocytes (TILs), demonstrating the potential of cell-based approaches in solid tumors. However, TIL therapy is just one of many strategies being explored. This review provides a comprehensive overview of the emerging field of immuno-oncology, focusing on how novel therapies targeting or harnessing components of the TME could enhance treatment efficacy and address persistent challenges in cancer care.
Collapse
Affiliation(s)
- Irena Barbara Padzińska-Pruszyńska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Bartłomiej Taciak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Łukasz Kiraga
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Anna Smolarska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Górczak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Paulina Kucharzewska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Kubiak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Jacek Szeliga
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, 65-046 Zielona Góra, Poland;
| | - Magdalena Król
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| |
Collapse
|
3
|
Ahuja S, Zaheer S. The evolution of cancer immunotherapy: a comprehensive review of its history and current perspectives. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2024; 20:51-73. [PMID: 39778508 PMCID: PMC11717579 DOI: 10.14216/kjco.24009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Cancer immunotherapy uses the body's immune system to combat cancer, marking a significant advancement in treatment. This review traces its evolution from the late 19th century to its current status. It began with William Coley's pioneering work using bacterial toxins to stimulate the immune system against cancer cells, establishing the foundational concept of immunotherapy. In the mid-20th century, cytokine therapies like interferons and interleukins emerged, demonstrating that altering the immune response could reduce tumors and highlighting the complex interplay between cancer and the immune system. The discovery of immune checkpoints, regulatory pathways that prevent autoimmunity but are exploited by cancer cells to evade detection, was a pivotal development. Another major breakthrough is CAR-T cell therapy, which involves modifying a patient's T cells to target cancer-specific antigens. This personalized treatment has shown remarkable success in certain blood cancers. Additionally, cancer vaccines aim to trigger immune responses against tumor-specific or associated antigens, and while challenging, ongoing research is improving their efficacy. The historical progression of cancer immunotherapy, from Coley's toxins to modern innovations like checkpoint inhibitors and CAR-T cell therapy, underscores its transformative impact on cancer treatment. As research delves deeper into the immune system's complexities, immunotherapy is poised to become even more crucial in oncology, offering renewed hope to patients globally.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| |
Collapse
|
4
|
Xu M, Pan Y. Chimeric Antigen Receptor (CAR)-T Cells: A New Era for Hepatocellular Carcinoma Treatment. J Biochem Mol Toxicol 2024; 38:e70091. [PMID: 39664011 DOI: 10.1002/jbt.70091] [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: 07/11/2024] [Revised: 09/24/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers and a worldwide health concern that requires novel treatment approaches. Tyrosine kinase inhibitors (TKIs) and immune checkpoint blockades (ICBs) are the current standard of care; however, their clinical benefits are limited in some advanced and metastatic patients. With the help of gene engineering techniques, a novel adoptive cellular therapy (ACT) called chimeric antigen receptor (CAR)-T cells was recently introduced for treating HCC. A plethora of current clinical and preclinical studies are attempting to improve the efficacy of CAR-T cells by dominating the immunosuppressive environment of HCC and finding the best tumor-specific antigens (TSAs). The future of care for HCC patients might be drastically improved due to the convergence of novel therapeutic methods and the continuous progress in ACT research. However, the clinical application of CAR-T cells in solid tumors is still facing several challenges. In this study, we provide an overview of the advancement and prospects of CAR-T cell immunotherapy in HCC, as well as an investigation of how cutting-edge engineering could improve CAR-T cell efficacy and safety profile.
Collapse
Affiliation(s)
- Ming Xu
- Department of Liver, Gallbladder, Spleen and Stomach, Heilongjiang Academy of Chinese Mediceal Sciences, Harbin, Heilongjiang, China
| | - Yang Pan
- Department of Liver, Gallbladder, Spleen and Stomach, Heilongjiang Academy of Chinese Mediceal Sciences, Harbin, Heilongjiang, China
| |
Collapse
|
5
|
He A, Li Q, Dang M, Lu W, Li X, Dai Z, Ding M, Zhang Y, Dong H, Teng Z, Mou Y. Extracellular Vesicle-Inspired Minimalist Flexible Nanocapsules Assembled with Whole Active Ingredients for Highly Efficient Enhancement of DC-Mediated Tumor Immunotherapy. Adv Healthc Mater 2024; 13:e2401199. [PMID: 39054675 PMCID: PMC11650550 DOI: 10.1002/adhm.202401199] [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: 03/31/2024] [Revised: 06/23/2024] [Indexed: 07/27/2024]
Abstract
The development of nanovaccines capable of eliciting tumor-specific immune responses holds significant promise for tumor immunotherapy. However, many nanovaccine designs rely heavily on incorporating multiple adjuvants and carriers, increasing the biological hazards associated with these additional components. Here, this work introduces novel flexible nanocapsules (OVAnano) designed to mimic extracellular vesicles, primarily using the ovalbumin antigen and minimal polyethylenimine adjuvant components. These results show that the biomimetic flexible structure of OVAnano facilitates enhanced antigen uptake by dendritic cells (DCs), leading to efficient antigen and adjuvant release into the cytosol via endosomal escape, and ultimately, successful antigen cross-presentation by DCs. Furthermore, OVAnano modulates the intracellular nuclear factor kappa-B (NF-κB) signaling pathway, promoting DC maturation. The highly purified antigens in OVAnano demonstrate remarkable antigen-specific immunogenicity, triggering strong antitumor immune responses mediated by DCs. Therapeutic tumor vaccination studies have also shown that OVAnano administration effectively suppresses tumor growth in mice by inducing immune responses from CD8+ and CD4+ T cells targeting specific antigens, reducing immunosuppression by regulatory T cells, and boosting the populations of effector memory T cells. These findings underscore that the simple yet potent strategy of employing minimal flexible nanocapsules markedly enhances DC-mediated antitumor immunotherapy, offering promising avenues for future clinical applications.
Collapse
Affiliation(s)
- Ao He
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolInstitute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| | - Qiang Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolInstitute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| | - Meng Dang
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for Biosensors, Institute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023China
| | - Wei Lu
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for Biosensors, Institute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023China
| | - Xiaoye Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolInstitute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| | - Zhuo Dai
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolInstitute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| | - Meng Ding
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolInstitute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| | - Yu Zhang
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolInstitute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| | - Heng Dong
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolInstitute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for Biosensors, Institute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023China
| | - Yongbin Mou
- Nanjing Stomatological HospitalAffiliated Hospital of Medical SchoolInstitute of StomatologyNanjing University30 Zhongyang RoadNanjingJiangsu210008China
| |
Collapse
|
6
|
Yang X, Liu Y, Wang Z, Jin Y, Gu W. Ferroptosis as a new tool for tumor suppression through lipid peroxidation. Commun Biol 2024; 7:1475. [PMID: 39521912 PMCID: PMC11550846 DOI: 10.1038/s42003-024-07180-8] [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: 06/11/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
As a newly defined type of programmed cell death, ferroptosis is considered a potent weapon against tumors due to its distinct mechanism from other types of programmed cell death. Ferroptosis is triggered by the uncontrolled accumulation of hydroperoxyl polyunsaturated fatty acid-containing phospholipids, also called lipid peroxidation. The lipid peroxidation, generated through enzymatic and non-enzymatic mechanisms, drives changes in cell morphology and the destruction of membrane integrity. Here, we dissect the mechanisms of ferroptosis induced enzymatically or non-enzymatically, summarize the major metabolism pathways in modulating lipid peroxidation, and provide insights into the relationship between ferroptosis and tumor suppression. In this review, we discuss the recent advances of ferroptosis in tumor microenvironments and the prospect of potential therapeutic application.
Collapse
Affiliation(s)
- Xin Yang
- Suzhou Ninth Hospital Affiliated to Soochow University, The Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| | - Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zhe Wang
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Ying Jin
- Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou Ninth People's Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
7
|
Xia HY, Zhao Y, Meng RY, Chen AZ, Wang SB, Kankala RK. Transition Metal (Molybdenum)-Doped Drug-like Conformational Nanoarchitectonics with Altered Valence States (Mn 2+/Mn 4+ and Mo 5+/Mo 6+) for Augmented Cancer Theranostics. Mol Pharm 2024; 21:5864-5879. [PMID: 39378327 DOI: 10.1021/acs.molpharmaceut.4c00896] [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: 10/10/2024]
Abstract
Despite the advancements in cancer therapy, delivering active pharmaceutical ingredients (APIs) using nanoparticles remains challenging due to the failed conveyance of the required drug payload, poor targeting ability, and poor biodistribution, hampering their clinical translation. Recently, the appropriate design of materials with intrinsic therapeutic functionalities has garnered enormous interest in the development of various intelligent therapeutic nanoplatforms. In this study, we demonstrate the fabrication of transition metal (molybdenum, Mo)-doped manganese dioxide (MnO2) nanoarchitectures, exhibiting diagnostic (magnetic resonance imaging, MRI) and therapeutic (chemodynamic therapy, CDT) functionalities. The facile hydrothermal approach-assisted Mo-doped MnO2 flower-like nanostructures offered tailorable morphologies in altered dimensions, precise therapeutic effects, exceptional biocompatibility, and biodegradability in the tumor microenvironment. The resultant defects due to doped Mo species exhibited peroxidase and oxidase activities, improving glutathione (GSH) oxidation. The two sets of variable valence metal ion pairs (Mn2+/Mn4+ and Mo5+/Mo6+) and their interplay could substantially improve the Fenton-like reaction and generate toxic hydroxyl radicals (•OH), thus achieving CDT-assisted antitumor effects. As inherent T1-MRI agents, these MnO2 nanoparticles displayed excellent MRI efficacy in vitro. Together, we believe that these conformational Mo-doped MnO2 nanoarchitectures with two pairs of variable valence states could potentiate drugless therapy in pharmaceutics.
Collapse
Affiliation(s)
- Hong-Ying Xia
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Ying Zhao
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Ruo-Yin Meng
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, P. R. China
| |
Collapse
|
8
|
Ma J, Hua L, Zhu Y, Mao G, Fu C, Qin S. Photo-Thermally Controllable Tumor Metabolic Modulation to Assist T Cell Activation for Boosting Immunotherapy. Int J Nanomedicine 2024; 19:11181-11194. [PMID: 39513087 PMCID: PMC11542477 DOI: 10.2147/ijn.s483815] [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: 06/22/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
Background Glycolysis is crucial for tumor cell proliferation, supporting their energy needs and influencing the tumor microenvironment (TME). On one hand, increased lactate levels produced by glycolysis acidifies the TME, inhibiting T cell activity. On the other hand, glycolysis promotes the expression of PD-L1 through various mechanisms, facilitating immune evasion. Therefore, controlled modulation of glycolysis in tumor cells to subsequently improve the immune tumor microenvironment holds significant implications for clinical cancer treatment and immune regulation. Methods To reverse the immunosuppressive microenvironment caused by tumor glycolysis and reduce tumor immune escape, we developed a photo-thermal-controlled precision drug delivery platform to regulate tumor metabolism and aid in the activation of T cells, thereby enhancing immunotherapy. First, hollow mesoporous Prussian blue (HPB) was prepared, and the glycolysis inhibitor 3-bromopyruvate (3-BrPA) was encapsulated within HPB using the phase-change material 1-tetradecanol, resulting in B/T-H. This product was then modified with tumor cell membranes to obtain a photo-thermal controllable regulator (B/T-H@Membrane, B/T-HM). Results Due to the excellent drug loading and photo-thermal properties of HPB, upon reaching the tumor, B/T-HM can rapidly heat under 808 nm irradiation, causing the 1-tetradecanol to transition to a liquid phase and release 3-BrPA, which effectively inhibits tumor glycolysis through the HK2 pathway, thereby reducing tumor cell proliferation, decreasing lactate production, and downregulating tumor PD-L1 expression. In synergy with photo-thermal and αPD-1, this photo-thermally controllable metabolic-immune therapy effectively activates T cells to eliminate tumor. Conclusion In response to the changes in immune microenvironment caused by tumor metabolism, a photo-thermal precision-controlled drug delivery platform was successfully developed. This platform reshapes the tumor immunosuppressive microenvironment, providing a new approach for T cell-based tumor immunotherapy. It also opens new avenues for photo-thermal controllable metabolic-immune therapy.
Collapse
Affiliation(s)
- Jun Ma
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Lixin Hua
- Department of General Surgery, Affiliated Huishan Hospital of Xinglin College, Nantong University, Wuxi Huishan District People’s Hospital, Wuxi, People’s Republic of China
| | - Yinxing Zhu
- Department of Traditional Chinese Medicines, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou, 225300, People’s Republic of China
| | - Guangyao Mao
- Institute of Clinical Medicine, the Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, People’s Republic of China
| | - Chunsheng Fu
- Institute of Clinical Medicine, the Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, People’s Republic of China
| | - Shiyue Qin
- Department of Ophthalmology, the Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, People’s Republic of China
| |
Collapse
|
9
|
Su X, Yu H, Lei Q, Chen X, Tong Y, Zhang Z, Yang W, Guo Y, Lin L. Systemic lupus erythematosus: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:54. [PMID: 39472388 PMCID: PMC11522254 DOI: 10.1186/s43556-024-00217-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disorder characterized by dysregulated immune responses and autoantibody production, which affects multiple organs and varies in clinical presentation and disease severity. The development of SLE is intricate, encompassing dysregulation within the immune system, a collapse of immunological tolerance, genetic susceptibilities to the disease, and a variety of environmental factors that can act as triggers. This review provides a comprehensive discussion of the pathogenesis and treatment strategies of SLE and focuses on the progress and status of traditional and emerging treatment strategies for SLE. Traditional treatment strategies for SLE have mainly employed non-specific approaches, including cytotoxic and immunosuppressive drugs, antimalarials, glucocorticoids, and NSAIDs. These strategies are effective in mitigating the effects of the disease, but they are not a complete cure and are often accompanied by adverse reactions. Emerging targeted therapeutic drugs, on the other hand, aim to control and treat SLE by targeting B and T cells, inhibiting their activation and function, as well as the abnormal activation of the immune system. A deeper understanding of the pathogenesis of SLE and the exploration of new targeted treatment strategies are essential to advance the treatment of this complex autoimmune disease.
Collapse
Affiliation(s)
- Xu Su
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Hui Yu
- Department of Urology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China
| | - Qingqiang Lei
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400000, China
| | - Xuerui Chen
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Yanli Tong
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, Paris, F-75015, France
| | - Zhongyang Zhang
- Department of Health Technology, The Danish National Research Foundation and Villum Foundation's Center IDUN, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Wenyong Yang
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Department of Neurosurgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China.
| | - Yuanbiao Guo
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| | - Liangbin Lin
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
10
|
Xu F, Ni Q, Gong N, Xia B, Zhang J, Guo W, Hu Z, Li J, Liang XJ. Delivery Systems Developed for Treatment Combinations to Improve Adoptive Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407525. [PMID: 39165065 DOI: 10.1002/adma.202407525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/26/2024] [Indexed: 08/22/2024]
Abstract
Adoptive cell therapy (ACT) has shown great success in the clinic for treating hematologic malignancies. However, solid tumor treatment with ACT monotherapy is still challenging, owing to insufficient expansion and rapid exhaustion of adoptive cells, tumor antigen downregulation/loss, and dense tumor extracellular matrix. Delivery strategies for combination cell therapy have great potential to overcome these hurdles. The delivery of vaccines, immune checkpoint inhibitors, cytokines, chemotherapeutics, and photothermal reagents in combination with adoptive cells, have been shown to improve the expansion/activation, decrease exhaustion, and promote the penetration of adoptive cells in solid tumors. Moreover, the delivery of nucleic acids to engineer immune cells directly in vivo holds promise to overcome many of the hurdles associated with the complex ex vivo cell engineering strategies. Here, these research advance, as well as the opportunities and challenges for integrating delivery technologies into cell therapy s are discussed, and the outlook for these emerging areas are criticlly analyzed.
Collapse
Affiliation(s)
- Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qiankun Ni
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Ningqiang Gong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Weisheng Guo
- College of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, 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, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
11
|
Xiong X, Wang X, Liu CC, Shao ZM, Yu KD. Deciphering breast cancer dynamics: insights from single-cell and spatial profiling in the multi-omics era. Biomark Res 2024; 12:107. [PMID: 39294728 PMCID: PMC11411917 DOI: 10.1186/s40364-024-00654-1] [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: 06/28/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024] Open
Abstract
As one of the most common tumors in women, the pathogenesis and tumor heterogeneity of breast cancer have long been the focal point of research, with the emergence of tumor metastasis and drug resistance posing persistent clinical challenges. The emergence of single-cell sequencing (SCS) technology has introduced novel approaches for gaining comprehensive insights into the biological behavior of malignant tumors. SCS is a high-throughput technology that has rapidly developed in the past decade, providing high-throughput molecular insights at the individual cell level. Furthermore, the advent of multitemporal point sampling and spatial omics also greatly enhances our understanding of cellular dynamics at both temporal and spatial levels. The paper provides a comprehensive overview of the historical development of SCS, and highlights the most recent advancements in utilizing SCS and spatial omics for breast cancer research. The findings from these studies will serve as valuable references for future advancements in basic research, clinical diagnosis, and treatment of breast cancer.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xin Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Luo Z, Zhu J, Fang Z, Xu R, Wan R, He Y, Chen Y, Chen S, Wang Q, Liu Q, Chen S. Exercise-augmented THSD7B exhibited a positive prognostic implication and tumor-suppressed functionality in pan-cancer. Front Immunol 2024; 15:1440226. [PMID: 39161765 PMCID: PMC11330788 DOI: 10.3389/fimmu.2024.1440226] [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: 05/29/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Background Breast cancer, one of the most prevalent malignancies among women worldwide, has rising incidence rates. Physical activity, particularly exercise, has emerged as a significant modifier of cancer prognosis, influencing both tumor biology and patient outcomes. Methods In this study, we utilized a murine breast cancer model, dividing mice into a control group and an exercise group; the latter underwent 21 days of voluntary running. We conducted RNA sequencing, bioinformatics analysis, pan-cancer analysis, and cellular experiments to investigate the underlying mechanisms influenced by exercise. Results Exercise led to a significant reduction in tumor size and weight. Post-exercise mRNA sequencing indicated a notable upregulation of THSD7B in the exercised mice, with significant alterations observed in pathways such as MicroRNAs in cancers and the Calcium signaling pathway. In a broader cancer context, THSD7B showed considerable expression variability, being significantly downregulated in several cancers, correlating with positive prognostic outcomes in PRAD, LAML, KIRC, and GBM and highlighting its potential role as a prognostic marker and therapeutic target. THSD7B expression was also negatively associated with processes of breast cancer cell proliferation, migration, and invasion. Conclusion This study underscores the dual role of exercise in modulating gene expression relevant to tumor growth and highlights the potential of THSD7B as a therapeutic target in cancer. Future research should further explore the specific mechanisms by which exercise and THSD7B influence cancer progression and develop immunotherapy-enhanced strategies to change patient outcomes in clinical settings.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinguo Zhu
- Department of Orthopaedics, Nantong Tongzhou Hospital of Traditional Chinese Medicine, Tongzhou, Jiangsu, China
| | - Zhengyuan Fang
- The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, Liaoning, China
| | - Rui Xu
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, China
| | - Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanwei He
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yisheng Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuo Chen
- Internal Medicine of Chinese Medicine, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Qing Wang
- Department of Orthopaedics, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu, China
| | - Qizhi Liu
- Internal Medicine of Chinese Medicine, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Nie S, Qin Y, Ou L, Chen X, Li L. In Situ Reprogramming of Immune Cells Using Synthetic Nanomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310168. [PMID: 38229527 DOI: 10.1002/adma.202310168] [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: 10/01/2023] [Revised: 01/12/2024] [Indexed: 01/18/2024]
Abstract
In the past decade, adoptive cell therapy with chimeric antigen receptor-T (CAR-T) cells has revolutionized cancer treatment. However, the complexity and high costs involved in manufacturing current adoptive cell therapy greatly inhibit its widespread availability and access. To address this, in situ cell therapy, which directly reprograms immune cells inside the body, has recently been developed as a promising alternative. Here, an overview of the recent progress in the development of synthetic nanomaterials is provided to deliver plasmid DNA or mRNA for in situ reprogramming of T cells and macrophages, focusing especially on in situ CAR therapies. Also, the main challenges for in situ immune cell reprogramming are discussed and some approaches to overcome these barriers to fulfill the clinical applications are proposed.
Collapse
Affiliation(s)
- Shihong Nie
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuyang Qin
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Liyuan Ou
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, 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
| | - Ling Li
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
14
|
Guo D, Huang Y, Wang K, Yang C, Ma L, Zhang Y, Yu H, Cui M, Tang Z. Preparation and Characterization Evaluation of Poly(L-Glutamic Acid)- g-Methoxy Poly(Ethylene Glycol)/Combretastatin A4/BLZ945 Nanoparticles for Cervical Cancer Therapy. Int J Nanomedicine 2023; 18:6901-6914. [PMID: 38026524 PMCID: PMC10676729 DOI: 10.2147/ijn.s441131] [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: 09/19/2023] [Accepted: 11/12/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Cervical cancer (CC) is a highly vascularized tumor with abundant abnormal blood vessel, which could be targeted by therapeutic strategies. Poly(L-glutamic acid)-g-methoxy poly(ethylene glycol)/combretastatin A4 (CA4)/BLZ945 nanoparticles (CB-NPs) have shown great potential as nano vascular disrupting agents (VDAs) in the realm of synergistic cancer therapy. Methods In this study, we investigated the nanocharacteristics of CB-NPs, focusing on active pharmaceutical ingredients (API), as well as lyophilized samples combining API with protective agents (PAs). The in vivo efficacy of final sample (API + PAs) was evaluated. Results The assembled sphere of API with complex core and thin-shell structure was confirmed. PAs were found to significantly influence in vivo efficacy. Collaborative efforts between API and PAs, namely mannitol and lactose, resulted in the most promising lyophilized sample, ie, the final sample (FS2) for CC therapy. Impressively, FS2 demonstrated an exceptional 100% cure rate on the CC U14-bearing mice model. Conclusion FS2 has provided significant insights for cervical cancer therapy. It is also crucial to develop a comprehensive evaluation strategy for the formulation of nanomedicine, which has the potential to serve as a guideline for future clinical trials.
Collapse
Affiliation(s)
- Dongmei Guo
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin Province, 130041, People’s Republic of China
| | - Yue Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Kun Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Chenguang Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Lili Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| | - Manhua Cui
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin Province, 130041, People’s Republic of China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People’s Republic of China
| |
Collapse
|