1
|
Wang N, Zhu M, Jiang Y, Zhang D, Lin J, Wu J, Lin N, Gao J. Neutrophil-Mediated Tumor Discrimination Biomimetic Nanodevice for Precise Tumor Eradication and Metastasis Cascade Perturbing. ACS APPLIED MATERIALS & INTERFACES 2025; 17:23680-23690. [PMID: 40219943 DOI: 10.1021/acsami.5c01866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
The deficient discrimination of tumor cells, limited accumulation of therapeutic agents in tumor foci, and undesirable tumor metastasis result in compromised therapeutic efficacy in antitumor therapy. Neutrophils, the most abundant circulating leukocytes, are key players in tumor progression and have a high affinity to tumors. Herein, a biomimetic tumor discrimination nanodevice (NL-FSB) is developed by harboring a pH-sensitive Fenton agent (FSB) in an activated neutrophil membrane-incorporated liposome for tumor-specific ablation. Inheriting the biointerfacing properties of neutrophils, NL-FSB is endowed with high affinity to tumors. On one hand, NL-FSB can be recruited to acidic tumor sites mediated by chemotaxis attraction and selectively generate reactive oxygen species via amplified Fenton chemical reaction for specific tumor eradication. On the other hand, the biomimetic NL-FSB can also target and bind tumor vascular endothelium or circulating tumor cells (CTCs) in circulation, executing pseudo escort to perturb CTC-neutrophil cluster formation and tumor metastasis cascade. Unprecedentedly, the neutrophil-mediated nanoagent can effectively inhibit the already-formed tumor and prevent tumor metastasis with high specificity. Our study represents a promising yet simple strategy for tumor-specific killing and metastasis cascade blockage via a nanobioengineering functionalization strategy.
Collapse
Affiliation(s)
- Nan Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Mingjian Zhu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yiming Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Daoming Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiachen Lin
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiahe Wu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Zheiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Jianqing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Zheiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
2
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [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: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
3
|
Cherfi I, Nasma M, Hasan GG, Benaissa A, Benaissa Y, Laouini SE, Bouafia A, Alharthi F, Emran TB, Mallick J. Therapeutic Potential of Artemisia campestris Essential Oil: Antioxidant, Anti-Inflammatory, and Anticancer Insights From In Silico Analysis. Biomed Chromatogr 2025; 39:e70012. [PMID: 39918166 DOI: 10.1002/bmc.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 03/04/2025]
Abstract
Artemisia campestris subsp. campestris (tuguft) is a medicinal plant traditionally used in Algerian medicine. This study investigates the chemical composition and bioactivity of its essential oil (ACEO). Gas chromatography-mass spectrometry (GC-MS) analysis identified key compounds, including linalyl acetate (2.92%), geranyl acetate (2.45%), and eucalyptol (1.38%). ACEO demonstrated significant antioxidant activity, with IC50 values of 11.09 μg/mL (DPPH), 15.81 μg/mL (FRAP), and 22.70 μg/mL (β-carotene). It also enhanced peroxidase activity by 82.67 U/g. The anti-inflammatory effects were confirmed with an IC50 of 18.87 μg/mL. Notably, in silico molecular docking revealed that 3-cyclopentyl-N-(2-(3,4-dimethoxyphenyl)ethyl) exhibits strong binding affinity to phosphoinositide 3-kinase gamma, a target for pancreatic cancer therapy, suggesting potential anticancer activity. These findings underscore the therapeutic potential of ACEO, highlighting its antioxidant, anti-inflammatory, and anticancer properties.
Collapse
Affiliation(s)
- Inasse Cherfi
- Faculty of Natural Science and Life, Department of Molecular and Cellular Biology, El Oued University, El Oued, Algeria
- Laboratory Biology, Environment, and Health, Faculty of Natural Sciences and Life, El-Oued University, El-Oued, Algeria
| | - Mahboub Nasma
- Faculty of Natural Science and Life, Department of Molecular and Cellular Biology, El Oued University, El Oued, Algeria
- Laboratory Biology, Environment, and Health, Faculty of Natural Sciences and Life, El-Oued University, El-Oued, Algeria
| | - Gamil Gamal Hasan
- VPRS Laboratory, Chemistry Department, Faculty of Mathematics and Matter Sciences, University of KASDI Merbah, Ouargla, Algeria
- Laboratory of Biotechnology Biomaterial and Condensed Matter, Faculty of Technology of Technology, University of El Oued, El Oued, Algeria
| | - Abir Benaissa
- Laboratory of Biotechnology Biomaterial and Condensed Matter, Faculty of Technology of Technology, University of El Oued, El Oued, Algeria
| | - Youcef Benaissa
- VPRS Laboratory, Chemistry Department, Faculty of Mathematics and Matter Sciences, University of KASDI Merbah, Ouargla, Algeria
| | - Salah Eddine Laouini
- Laboratory of Biotechnology Biomaterial and Condensed Matter, Faculty of Technology of Technology, University of El Oued, El Oued, Algeria
- Department of Process Engineering, Faculty of Technology, University of El Oued, El-Oued, Algeria
| | - Abderrhmane Bouafia
- Laboratory of Biotechnology Biomaterial and Condensed Matter, Faculty of Technology of Technology, University of El Oued, El Oued, Algeria
- Department of Process Engineering, Faculty of Technology, University of El Oued, El-Oued, Algeria
| | - Fahad Alharthi
- Department of Chemistry, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Talha Bin Emran
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center, Brown University, Providence, Rhode Island, USA
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Jewel Mallick
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| |
Collapse
|
4
|
Nisar MF, Yan T, Cai Y, Wan C. Immuno-oncological Challenges and Chemoresistance in Veterinary Medicine: Probiotics as a New Strategic Tool. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10468-8. [PMID: 39954194 DOI: 10.1007/s12602-025-10468-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/17/2025]
Abstract
Cancer has the highest death rates due to increased immuno-oncological (IO) challenges and chemoresistance caused by gut dysbiosis, whereas administration of probiotics may reverse these responses against anticancer therapies. Recently, immunotherapeutics have extensively been focused for significant advancements in pharmacological drug discovery and clinical outcomes. Mammals have intestinal epithelial cells, mucosal immune cells, and indigenous gut microbiota which may reshape immunotherapeutics efficacy. These include use of T-cell immune checkpoint inhibitors (ICPI), genetically engineered T-cells, tumor vaccines, monoclonal antibodies (mAbs), and anti-B- and T-cell antibodies. Immunotherapeutics for cancer treatment became popular in both veterinary and human health care systems due to their strong inhibitory actions against PD-1 and CTLA-4 to check tumorigenesis. IO issues in animals also need special attention, where caninized mAbs targeting CD-20 and CD-52 have been clinically used in treating canine B-cell and T-cell lymphomas, respectively. Probiotics appeared as strong immunotherapeutics that might be shaping the epigenetics of the organisms specifically in animal breeding practices for desired features, but limited literature regarding the immunomodulatory effects in humans and animals is available. In addition, considering the important role of probiotics in humans and veterinary medicine, a new perspective on the probiotic-mediated modulation of ncRNAs (miRNAs, lncRNAs, circRNAs) is also highlighted and would be a new therapeutic tool. This review provides insight into the cellular processes and pharmacological activities for treating veterinary infectious diseases and covers small drug molecules as ncRNA-modulators in veterinary medicine.
Collapse
Affiliation(s)
- Muhammad Farrukh Nisar
- Ministry of Education and Jiangxi Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Jiangxi Agricultural University, Nanchang, 330045, China
- Jiangxi Key Laboratory for Post-harvest Technology and Nondestructive Testing of Fruits & Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, China
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, Pakistan
| | - Tingdong Yan
- School of Pharmacy, Nantong University, Nantong, 226001, China.
| | - Yi Cai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Chunpeng Wan
- Jiangxi Key Laboratory for Post-harvest Technology and Nondestructive Testing of Fruits & Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
5
|
Jiang X, Lin W. Innate Immune Activation with Multifunctional Nanoparticles for Cancer Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202423280. [PMID: 39752565 DOI: 10.1002/anie.202423280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Immune checkpoint blockade (ICB) has revolutionized the treatment of many cancers by leveraging the immune system to combat malignancies. However, its efficacy is limited by the immunosuppressive tumor microenvironment and other regulatory mechanisms of the immune system. Innate immune modulators (IIMs) provide potent immune activation to complement adaptive immune responses and help overcome resistance to ICB. This minireview provides an overview of IIMs and their roles in antitumor immune responses and summarizes recent advances in developing nanotechnology to enhance the delivery of IIMs to tumors for potentiating cancer immunotherapy and mitigating systemic toxicity. We discuss innovative nanoparticle platforms for the delivery of IIMs targeting the cyclic GMP-AMP synthase-stimulator of interferon genes pathway, the toll-like receptor pathway, and the retinoic acid-inducible gene I-like receptor pathway. We review the preliminary clinical readouts of representative IIM nanoptherapeutics and highlight the development of multifunctional nanoparticles for combination treatments of IIMs with conventional treatment mdoalities such as chemotherapy, radiotherapy, photodynmic therapy, and tumor antigens. Finally, we summarize the lessons learned from the existing systems, the challenges in the field, and future perspectives for this exciting field of nanotherapeutics for cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaomin Jiang
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
6
|
Lin Y, Jin H, She Y, Zhang Y, Cui L, Xie C, Liu Y, Zhang H, Guo H, Wu J, Li L, Guo Z, Wang X, Jiang W, Chen X, He S, Zhou P, Tan J, Bei JX, Liu J, Chen YX, Zhao Q, Xia X, Wang Z. CBX2 suppresses interferon signaling to diminish tumor immunogenicity via a noncanonical corepressor complex. Proc Natl Acad Sci U S A 2025; 122:e2417529122. [PMID: 39883845 PMCID: PMC11804501 DOI: 10.1073/pnas.2417529122] [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: 08/28/2024] [Accepted: 12/27/2024] [Indexed: 02/01/2025] Open
Abstract
Chromobox 2 (CBX2), a crucial component of the polycomb repressive complex (PRC), has been implicated in the development of various human cancers. However, its role in the regulation of tumor immunogenicity and immune evasion remains inadequately understood. In this study, we found that ablation of CBX2 led to tumor growth inhibition, activation of the tumor immune microenvironment, and enhanced therapeutic efficacy of anti-PD1 or adoptive T cell therapies by using murine syngeneic tumor models. By analysis of the CBX2-regulated transcriptional program coupled with mass spectrometry screening of CBX2-interacting proteins, we found that CBX2 suppresses interferon signaling independent of its function in the canonical PRC. Mechanistically, CBX2 directly interacts with RACK1 and facilitates the recruitment of HDAC1, which attenuates the H3K27ac modification on the promoter regions of interferon-stimulated genes, thereby suppressing interferon signaling. Consequently, CBX2 reduces tumor immunogenicity and enables immune evasion. Moreover, a high expression level of CBX2 is associated with immune suppressive tumor microenvironment and reduced efficacy of immunotherapy across various human cancer types. Our study identifies a noncanonical CBX2-RACK1-HDAC1 corepressor complex in suppression of tumor immunogenicity, thereby presenting a potential target and biomarker for tumor immunotherapy.
Collapse
Affiliation(s)
- Yanxun Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Huan Jin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Yong She
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Yiqun Zhang
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Lei Cui
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Chunyuan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Yongxiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Huanling Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Hui Guo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Jiaxin Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Lin Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Zixuan Guo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Xiaojuan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Wu Jiang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Xu Chen
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou510080, China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Penghui Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Jing Tan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Jinyun Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
- Platform of Metabolomics Center for Precision Medicine, Metabolic Innovation Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou510080, China
| | - Yan-Xing Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Qi Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
- Hainan Academy of Medical Sciences, Hainan Medical University, Haikou571199, China
| | - Zining Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510050, China
| |
Collapse
|
7
|
Bogani G, Moore KN, Ray-Coquard I, Lorusso D, Matulonis UA, Ledermann JA, González-Martín A, Kurtz JE, Pujade-Lauraine E, Scambia G, Caruso G, Raspagliesi F, Colombo N, Monk BJ. Incorporating immune checkpoint inhibitors in epithelial ovarian cancer. Gynecol Oncol 2025; 193:30-40. [PMID: 39764856 DOI: 10.1016/j.ygyno.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/08/2024] [Accepted: 12/18/2024] [Indexed: 03/03/2025]
Abstract
OBJECTIVE Therapeutic interventions for epithelial ovarian cancer (EOC) have increased greatly over the last decade but improvements outside of biomarker selected therapies have been limited. There remains a pressing need for more effective treatment options that can prolong survival and enhance the quality of life of patients with EOC. In contrast to the significant benefits of immunotherapy with immune checkpoint inhibitors (CPI) seen in many solid tumors, initial experience in EOC suggests limited efficacy of CPIs monotherapy. METHODS A systematic review of phase III studies testing the role of CPIs in ovarian cancer was performed. RESULTS Seven randomized trials testing CPIs in newly diagnosed (n = 3) and recurrent (n = 4) EOC are evaluated. Overall, those trials included data of 5671 patients. Single-agent PD-L1 inhibitor trials have not shown significant efficacy in newly diagnosed ovarian cancer. Triplet maintenance with bevacizumab plus olaparib and durvalumab is associated with longer progression-free survival than maintenance with bevacizumab alone in patients without tumor BRCA mutations. CPIs were not effective in platinum-sensitive (n = 1031) and platinum-resistant (n = 1420) EOC. CONCLUSIONS The value of adding CPI to standard treatment including poly (ADP-ribose) polymerase (PARP) inhibitors with or without bevacizumab remains unclear and is being addressed in ongoing clinical trials. The combination of cytotoxic T-lymphocyte associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) inhibitors may enhance the efficacy of immunotherapy in EOC and studies are underway to investigate the combination of CPI with other emerging treatment modalities. PROSPERO registration ID: CRD42024536017.
Collapse
Affiliation(s)
- Giorgio Bogani
- Deaprtment of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.
| | - Kathleen N Moore
- Stephenson Cancer Center at the University of Oklahoma Health Sciences Center/Sarah Cannon Research Institute, Oklahoma City, OK, United States.
| | - Isabelle Ray-Coquard
- Centre Anticancereux Léon Bérard, University Claude Bernard Lyon, GINECO Group, Lyon, France.
| | - Domenica Lorusso
- Department of Gynecologic Oncology, Humanitas San Pio X, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
| | - Ursula A Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America.
| | | | - Antonio González-Martín
- Medical Oncology Department, Translational Oncology Group, CIMA, Universidad de Navarra, Cancer Center Clinica Universidad de Navarra, Madrid, and Grupo Español de Investigación en Cáncer ginecológicO (GEICO), Madrid, Spain.
| | - Jean-Emmanuel Kurtz
- Department of Medical and Surgical Oncology & Hematology, ICANS, Strasbourg, France.
| | - Eric Pujade-Lauraine
- Association de Recherche Cancers Gynécologiques (ARCAGY)-Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Paris, France; GINECO, Paris, France.
| | - Giovanni Scambia
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy.
| | - Giuseppe Caruso
- Department of Gynecology, European Institute of Oncology, IEO, IRCCS, Milan, Italy; Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| | - Francesco Raspagliesi
- Deaprtment of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.
| | - Nicoletta Colombo
- Gynecology Program, European Institute of Oncology, IEO, IRCCS, Milan, Italy; Department of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy.
| | - Bradley J Monk
- GOG Foundation, Florida Cancer Specialists and Research Institute, West Palm Beach, FL 33401, United States of America.
| |
Collapse
|
8
|
Meena SS, Kosgei BK, Soko GF, Tingjun C, Chambuso R, Mwaiselage J, Han RPS. Developing anti-TDE vaccine for sensitizing cancer cells to treatment and metastasis control. NPJ Vaccines 2025; 10:18. [PMID: 39870669 PMCID: PMC11772600 DOI: 10.1038/s41541-024-01035-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 11/21/2024] [Indexed: 01/29/2025] Open
Abstract
Tumor-derived exosomes (TDEs) mediate oncogenic communication, which modifies target cells to reinforce a tumor-promoting microenvironment. TDEs support cancer progression by suppressing anti-tumor immune responses, promoting metastasis, and conferring drug resistance. Thus, targeting TDEs could improve the efficacy of anti-cancer treatments and control metastasis. Current strategies to inhibit TDE-mediated oncogenic communication including drug-based and genetic modification-based inhibition of TDE release and/or uptake, have proved to be inefficient. In this work, we propose TDE surface engineering to express foreign antigens that will trigger life-long anti-TDE immune responses. The possibility of combining the anti-TDE vaccines with other treatments such as chemotherapy, radiotherapy, targeted therapy, and surgery is also explored.
Collapse
Affiliation(s)
- Stephene S Meena
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
- Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
- Ocean Road Cancer Institute, Dar es Salaam, United Republic of Tanzania.
| | - Benson K Kosgei
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Geofrey F Soko
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Ocean Road Cancer Institute, Dar es Salaam, United Republic of Tanzania
| | - Cheng Tingjun
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Ramadhani Chambuso
- Department of Global Health and Population, Harvard Chan School of Public Health, Harvard University, Cambridge, MA, USA
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Julius Mwaiselage
- Ocean Road Cancer Institute, Dar es Salaam, United Republic of Tanzania
| | - Ray P S Han
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
- Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China.
| |
Collapse
|
9
|
Ghaneialvar H, Jahani S, Hashemi E, Khalilzad MA, Falahi S, Rashidi MA, Majidpoor J, Najafi S. Combining anti-checkpoint immunotherapies and cancer vaccines as a novel strategy in oncological therapy: A review. Hum Immunol 2025; 86:111209. [PMID: 39662393 DOI: 10.1016/j.humimm.2024.111209] [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: 07/24/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
The field of cancer immunotherapy has experienced remarkable advancements in the treatment of human cancers over recent decades. Therapeutic cancer vaccines have been employed to elicit antitumor immune responses through the generation of specific reactions against tumor-associated antigens. Although preclinical studies have demonstrated hopeful results and at least one product is approved for clinical use, the overall efficacy of cancer vaccines remains restricted. The co-administration of anti-checkpoint antibodies alongside cancer vaccines is proposed as a potential strategy to enhance the clinical efficacy of immunotherapies. Among the various anti-checkpoint agents, monoclonal antibodies targeting CD127, OX40, and CD40 have been further investigated in combined administration with cancer vaccines, demonstrating a synergistic impact on disease outcomes in both animal models and human subjects. This combinational approach has been shown to suppress tumor regression, improve survival rates, and promote the efficacy of cancer vaccines via multiple mechanisms, including the augmentation of generation, activation, and expansion of CD8+ T cells, as well as the production of tumor-inhibitory cytokines. Importantly, the impact of the concurrent administration of anti-checkpoint agents and cancer vaccines surpass those observed with the sole vaccine, indicating that this strategy may offer significant advantages for clinical application in cancer patients. In this review, we aim to provide a comprehensive overview of the significance and therapeutic potential of the combined administration of checkpoint agonist/antagonist antibodies and cancer vaccines for human tumors.
Collapse
Affiliation(s)
- Hori Ghaneialvar
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Saleheh Jahani
- Department of Pathology, School of Medicine, University of California, San Diego, USA
| | - Elham Hashemi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Shahab Falahi
- Zoonotic Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Mohammad Amin Rashidi
- Department of Occupational Health and Safety, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran.
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Luo D, Zhong Q, Yue H, Wang J, Liang Q, Liu W, Zhu X. The predictors of lymphopenia and its effects on survival in locally advanced esophageal squamous cell carcinoma. Cancer Biol Ther 2024; 25:2371632. [PMID: 38946404 PMCID: PMC11218796 DOI: 10.1080/15384047.2024.2371632] [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: 12/15/2022] [Accepted: 06/19/2024] [Indexed: 07/02/2024] Open
Abstract
To investigate the impact of the effective radiation dose to immune cells (EDIC) and gross tumor volume (GTV) on lymphopenia and survival in patients with locally advanced esophageal squamous cell carcinoma (LAESCC). Between January 2013 and December 2020, 272 LAESCC patients were treated with definitive radiotherapy in two institutions. Based on radiation doses to the lungs, heart, and body region scanned, EDIC was calculated as an equal uniform dose to the total blood considering blood flow and fraction effect. The radiotherapy plan was used to calculate the GTVs. Lymphopenia was graded based on the lowest lymphocyte count during RT. The overall survival (OS), progress-free survival (PFS), and local recurrence-free survival (LRFS) were analyzed statistically. The lowest lymphocyte count was significantly correlated with EDIC (r= -0.389, p < .001) and GTV (r= -0.211, p < .001). Lymphopenia, EDIC, and GTV are risk factors for patients with ESCC. In a Kaplan-Meier analysis with EDIC and GTV as stratification factors, lymphopenia was not associated with OS in the EDIC>12.9 Gy group (p = .294)and EDIC ≤ 12.9 Gy group, and it was also not associated with OS in GTV>68.8 cm3 group (p = .242) and GTV ≤ 68.8 cm3 group(p = .165). GTV and EDIC had an impact on the relationship between lymphopenia and OS in patients with LAESCC undergoing definitive RT. Poorer OS, PFS, and LRFS are correlated with lymphopenia, higher EDIC, and larger GTV.
Collapse
Affiliation(s)
- Danjing Luo
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiulu Zhong
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haiying Yue
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jue Wang
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qianfu Liang
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenqi Liu
- Department of Radiation Oncology, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaodong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
- Department of Oncology, Wuming Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
11
|
Abdelfattah NS, Kula T, Elledge SJ. T-Switch: A specificity-based engineering platform for developing safe and effective T cell therapeutics. Immunity 2024; 57:2945-2958.e5. [PMID: 39631392 DOI: 10.1016/j.immuni.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 10/15/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
Many promising targets for adoptive T cell therapy (ACT) are self-antigens, but self-reactive T cells are generally eliminated during thymic selection or diverted to regulatory phenotypes. To bypass T cell tolerance and obtain potent and safe T cell therapeutics, we developed T-Switch, an in vitro T cell receptor (TCR) engineering platform for the creation, modification, and comprehensive profiling of TCRs that can target self-antigens. T-Switch first expands T cells that recognize a "foreign" peptide closely related to a self-antigen. The fine specificity of the TCR is then modified by directed evolution of the peptide binding region to switch its specificity to the self-antigen of interest. We applied T-Switch to engineer synthetic TCRs reactive to a tumor-associated self-antigen, validated the safety and efficacy of this approach, and detected no off-target recognition as measured against the human proteome. Thus, T-Switch represents a resource for the creation of collections of highly sensitive synthetic TCRs for T cell-based immunotherapies.
Collapse
Affiliation(s)
- Nouran S Abdelfattah
- Department of Genetics, Harvard Medical School, Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Tomasz Kula
- Department of Genetics, Harvard Medical School, Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Stephen J Elledge
- Department of Genetics, Harvard Medical School, Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Feng Z, Wang Y, Liang Y, Gu X, Yang Y, Zhang Y, Peng Q. Development and validation of a prognostic risk score model for hepatocellular carcinoma in the Asian population based on immunogenic cell death-related genes. Discov Oncol 2024; 15:744. [PMID: 39630208 PMCID: PMC11618282 DOI: 10.1007/s12672-024-01630-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/25/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), the predominant form of liver cancer, is marked by limited therapeutic success and unfavorable prognoses. Its etiology varies regionally, with hepatitis B virus (HBV) being the predominant cause in most of Asia. Immunogenic cell death (ICD), a specific type of cell death, has been extensively linked to HCC treatment in numerous studies. This research aims to explore the significance of ICD-related genes in the Asian HCC cohort, potentially offering novel approaches for HCC management. METHODS We initially obtained transcriptomic and clinical data pertinent to Asian HCC from the TCGA database. Subsequently, we classified the samples into distinct subgroups according to ICD gene expression levels and conducted analyses of the tumor microenvironment and enrichment. Furthermore, we randomly allocated the samples into training and testing cohorts, thereafter developing and validating an ICD gene-based prognostic model tailored for the Asian HCC population. RESULTS The Asian HCC samples were categorized into two subgroups: high and low ICD expression. In the low ICD expression group, we observed diminished infiltration of immune and stromal cells, increased tumor purity, and improved prognosis. Moreover, we devised a 5-gene risk-score prognostic model comprising BAX, CASP8, HMGB1, HSP90AA1, and IL6, demonstrating efficacy in prognostic predictions for the Asian HCC cohort. CONCLUSION Our investigation unveils new perspectives on the influence of ICDs within Asian HCC populations. The derived 5-gene risk-score prognostic model, based on ICDs, not only serves as a tool for assessing prognosis in Asian HCC cases but also suggests potential therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Zhengyang Feng
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Yanjie Wang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Liang
- Department of Oncology, The Fifth People's Hospital of Huai'an, Huai'an, China
| | - Xuhao Gu
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yinyin Yang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yusong Zhang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Qiliang Peng
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
13
|
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
|
14
|
Albertí-Valls M, Olave S, Olomí A, Macià A, Eritja N. Advances in Immunotherapy for Endometrial Cancer: Insights into MMR Status and Tumor Microenvironment. Cancers (Basel) 2024; 16:3918. [PMID: 39682106 DOI: 10.3390/cancers16233918] [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: 10/30/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Endometrial cancer is one of the most common gynecological malignancies, and while early-stage cases are highly treatable, recurrent or advanced EC remains challenging to manage. Immunotherapy, particularly immune checkpoint inhibitors, has revolutionized treatment approaches in oncology, and its application in EC has shown promising results. Key to immunotherapy efficacy in EC is the tumor's mismatch repair status, with MMR-deficient tumors demonstrating a higher tumor mutational burden and increased PD-L1 expression, making them more susceptible to immune checkpoint inhibitors (ICIs) such as pembrolizumab, durvalumab, and dostarlimab. However, not all mismatch repair-deficient (MMRd) tumors respond to ICIs, particularly those with a "cold" tumor microenvironment (TME) characterized by poor immune infiltration. In contrast, some MMR-proficient tumors with a "hot" TME respond well to ICIs, underscoring the complex interplay between MMR status, tumor mutational burden (TMB), and TME. To overcome resistance in cold tumors, novel therapies, including Chimeric Antigen Receptor (CAR) T cells and tumor-infiltrating lymphocytes are being explored, offering targeted immune-based strategies to enhance treatment efficacy. This review discusses the current understanding of immunotherapy in EC, emphasizing the prognostic and therapeutic implications of MMR status, TME composition, and emerging cell-based therapies.
Collapse
Affiliation(s)
- Manel Albertí-Valls
- Oncologic Pathology Group, Biomedical Research Institute of Lleida (IRBLleida), University of Lleida (UdL), Av. Rovira Roure 80, 25198 Lleida, Spain
| | - Sara Olave
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Anna Olomí
- Developmental and Oncogenic Signaling, Biomedical Research Institute of Lleida (IRBLleida), University of Lleida (UdL), Av. Rovira Roure 80, 25198 Lleida, Spain
| | - Anna Macià
- Oncologic Pathology Group, Biomedical Research Institute of Lleida (IRBLleida), University of Lleida (UdL), Av. Rovira Roure 80, 25198 Lleida, Spain
| | - Núria Eritja
- Oncologic Pathology Group, Biomedical Research Institute of Lleida (IRBLleida), University of Lleida (UdL), Av. Rovira Roure 80, 25198 Lleida, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
15
|
Liu Y, Deng Y, Yang C, Naranmandura H. Double-Faced Immunological Effects of CDK4/6 Inhibitors on Cancer Treatment: Challenges and Perspectives. Bioengineering (Basel) 2024; 11:1084. [PMID: 39593745 PMCID: PMC11591775 DOI: 10.3390/bioengineering11111084] [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: 10/04/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Cyclin-dependent kinases (CDKs) are generally involved in the progression of cell cycle and cell division in normal cells, while abnormal activations of CDKs are deemed to be a driving force for accelerating cell proliferation and tumorigenesis. Therefore, CDKs have become ideal therapeutic targets for cancer treatment. The U.S FDA has approved three CDK4/6 inhibitors (CDK4/6is) for the treatment of patients with hormone receptor-positive (HR+) or human epidermal growth factor receptor 2-negative (HER2-) advanced or metastatic breast cancer, and these drugs showed impressive results in clinics. Besides cell-cycle arrest, there is growing evidence that CDK4/6is exert paradoxical roles on cancer treatment by altering the immune system. Indeed, clinical data showed that CDK4/6is could change the immune system to exert antitumor effects, while these changes also caused tumor resistance to CDK4/6i. However, the molecular mechanism for the regulation of the immune system by CDK4/6is is unclear. In this review, we comprehensively discuss the paradoxical immunological effects of CDK4/6is in cancer treatment, elucidating their anticancer mechanisms through immunomodulatory activity and induction of acquired drug resistance by dysregulating the immune microenvironment. More importantly, we suggest a few strategies including combining CDK4/6is with immunotherapy to overcome drug resistance.
Collapse
Affiliation(s)
- Yongqin Liu
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yiying Deng
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chang Yang
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hua Naranmandura
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
16
|
Pergolizzi RG, Brower ST. Molecular Targets for the Diagnosis and Treatment of Pancreatic Cancer. Int J Mol Sci 2024; 25:10843. [PMID: 39409171 PMCID: PMC11476914 DOI: 10.3390/ijms251910843] [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: 08/07/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Pancreatic cancer is one of the most aggressive and lethal forms of cancer, with a five-year survival rate of less than 10%. Despite advances in treatment modalities, the prognosis for pancreatic cancer patients remains poor, highlighting the urgent need for innovative approaches for early diagnosis and targeted therapies. In recent years, there has been significant progress in understanding the molecular mechanisms underlying pancreatic cancer development and progression. This paper reviews the current knowledge of molecular targets for the diagnosis and treatment of pancreatic cancer.
Collapse
Affiliation(s)
| | - Steven T. Brower
- Department of Surgical Oncology and HPB Surgery, Englewood Health, Englewood, NJ 07631, USA
| |
Collapse
|
17
|
Lv M, Liu B, Duan Y, Lin J, Dai L, Li Y, Yu J, Liao J, Zhang J, Duan Y. Engineered Biomimetic Nanovesicles Synergistically Remodel Folate-Nucleotide and γ-Aminobutyric Acid Metabolism to Overcome Sunitinib-Resistant Renal Cell Carcinoma. ACS NANO 2024; 18:27487-27502. [PMID: 39329191 DOI: 10.1021/acsnano.4c08055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Reprogramming of cellular metabolism in tumors promoted the epithelial-mesenchymal transition (EMT) process and established immune-suppressive tumor microenvironments (iTME), leading to drug resistance and tumor progression. Therefore, remodeling the cellular metabolism of tumor cells was a promising strategy to overcome drug-resistant tumors. Herein, CD276 and MTHFD2 were identified as a specific marker and a therapeutic target, respectively, for targeting sunitinib-resistant clear cell renal cell carcinoma (ccRCC) and its cancer stem cell (CSC) population. The blockade of MTHFD2 was confirmed to overcome drug resistance via remodeling of folate-nucleotide metabolism. Moreover, the manganese dioxide nanoparticle was proven here by a high-throughput metabolome to be capable of remodeling γ-aminobutyric acid (GABA) metabolism in tumor cells to reconstruct the iTME. Based on these findings, engineered CD276-CD133 dual-targeting biomimetic nanovesicle EMφ-siMTHFD2-MnO2@Suni was designed to overcome drug resistance and terminate tumor progression of ccRCC. Using ccRCC-bearing immune-humanized NPG model mice, EMφ-siMTHFD2-MnO2@Suni was observed to remodel folate-nucleotide and GABA metabolism to deactivate the EMT process and reconstruct the iTME thereby overcoming the drug resistance. In the incomplete-tumor-resection recurrence model and metastasis model, EMφ-siMTHFD2-MnO2@Suni reduced recurrence and metastasis in vivo. This work thus provided an innovative approach that held great potential in the treatment of drug-resistant ccRCC by remodeling cellular metabolism.
Collapse
Affiliation(s)
- Minchao Lv
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2200/25 Xietu Rd, Shanghai 200032, China
| | - Bin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2200/25 Xietu Rd, Shanghai 200032, China
| | - Yi Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2200/25 Xietu Rd, Shanghai 200032, China
| | - Jiangtao Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2200/25 Xietu Rd, Shanghai 200032, China
| | - Li Dai
- Department of Otolaryngology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd, Shanghai 200127, China
| | - Yuanyuan Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2200/25 Xietu Rd, Shanghai 200032, China
| | - Jian Yu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2200/25 Xietu Rd, Shanghai 200032, China
| | - Jinghan Liao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2200/25 Xietu Rd, Shanghai 200032, China
| | - Jiali Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2200/25 Xietu Rd, Shanghai 200032, China
| | - Yourong Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 2200/25 Xietu Rd, Shanghai 200032, China
| |
Collapse
|
18
|
Parashar D, Mukherjee T, Gupta S, Kumar U, Das K. MicroRNAs in extracellular vesicles: A potential role in cancer progression. Cell Signal 2024; 121:111263. [PMID: 38897529 DOI: 10.1016/j.cellsig.2024.111263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
Intercellular communication, an essential biological process in multicellular organisms, is mediated by direct cell-to-cell contact and cell secretary molecules. Emerging evidence identifies a third mechanism of intercellular communication- the release of extracellular vesicles (EVs). EVs are membrane-enclosed nanosized bodies, released from cells into the extracellular environment, often found in all biofluids. The growing body of research indicates that EVs carry bioactive molecules in the form of proteins, DNA, RNAs, microRNAs (miRNAs), lipids, metabolites, etc., and upon transferring them, alter the phenotypes of the target recipient cells. Interestingly, the abundance of EVs is found to be significantly higher in different diseased conditions, most importantly cancer. In the past few decades, numerous studies have identified EV miRNAs as an important contributor in the pathogenesis of different types of cancer. However, the underlying mechanism behind EV miRNA-associated cancer progression and how it could be used as a targeted therapy remain ill-defined. The present review highlights how EV miRNAs influence essential processes in cancer, such as growth, proliferation, metastasis, angiogenesis, apoptosis, stemness, immune evasion, resistance to therapy, etc. A special emphasis has been given to the potential role of EV miRNAs as cancer biomarkers. The final section of the review delineates the ongoing clinical trials on the role of miRNAs in the progression of different types of cancer. Targeting EV miRNAs could be a potential therapeutic means in the treatment of different forms of cancer alongside conventional therapeutic approaches.
Collapse
Affiliation(s)
- Deepak Parashar
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Tanmoy Mukherjee
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA.
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Umesh Kumar
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad 201015, Uttar Pradesh, India.
| | - Kaushik Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani 741251, West Bengal, India.
| |
Collapse
|
19
|
Bianca C. A decade of thermostatted kinetic theory models for complex active matter living systems. Phys Life Rev 2024; 50:72-97. [PMID: 39002422 DOI: 10.1016/j.plrev.2024.06.015] [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] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
In the last decade, the thermostatted kinetic theory has been proposed as a general paradigm for the modeling of complex systems of the active matter and, in particular, in biology. Homogeneous and inhomogeneous frameworks of the thermostatted kinetic theory have been employed for modeling phenomena that are the result of interactions among the elements, called active particles, composing the system. Functional subsystems contain heterogeneous active particles that are able to perform the same task, called activity. Active matter living systems usually operate out-of-equilibrium; accordingly, a mathematical thermostat is introduced in order to regulate the fluctuations of the activity of particles. The time evolution of the functional subsystems is obtained by introducing the conservative and the nonconservative interactions which represent activity-transition, natural birth/death, induced proliferation/destruction, and mutation of the active particles. This review paper is divided in two parts: In the first part the review deals with the mathematical frameworks of the thermostatted kinetic theory that can be found in the literature of the last decade and a unified approach is proposed; the second part of the review is devoted to the specific mathematical models derived within the thermostatted kinetic theory presented in the last decade for complex biological systems, such as wound healing diseases, the recognition process and the learning dynamics of the human immune system, the hiding-learning dynamics and the immunoediting process occurring during the cancer-immune system competition. Future research perspectives are discussed from the theoretical and application viewpoints, which suggest the important interplay among the different scholars of the applied sciences and the desire of a multidisciplinary approach or rather a theory for the modeling of every active matter system.
Collapse
Affiliation(s)
- Carlo Bianca
- EFREI Research Lab, Université Paris-Panthéon-Assas, 30/32 Avenue de la République, 94800 Villejuif, France.
| |
Collapse
|
20
|
Luna OF, Perez YV, Ferrari DP, Sayedipour SS, Royo M, Acosta GA, Cruz LJ, Alves F, Agner E, Sydnes MO, Albericio F. Impact of N-Terminal PEGylation on Synthesis and Purification of Peptide-Based Cancer Epitopes for Pancreatic Ductal Adenocarcinoma (PDAC). ACS OMEGA 2024; 9:34544-34554. [PMID: 39157077 PMCID: PMC11325526 DOI: 10.1021/acsomega.4c02604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/30/2024] [Accepted: 06/10/2024] [Indexed: 08/20/2024]
Abstract
Peptide-based cancer vaccines have shown promising results in preclinical trials focusing on tumor immunotherapy. However, the presence of hydrophobic amino acid segments within these peptide sequences poses challenges in their synthesis, purification, and solubility, thereby hindering their potential use as cancer vaccines. In this study, we successfully synthesized peptide sequences derived from mesothelin (MSLN), a tumor-associated antigen overexpressed in pancreatic ductal adenocarcinoma (PDAC) by conjugating them with monodisperse polyethylene glycol (PEG). By PEGylating mesothelin epitopes of varying lengths (ranging from 9 to 38 amino acids) and hydrophobicity (60-90%), we achieved an effective method to improve the peptide yield and facilitate the processes of synthesis and purification. PEGylation significantly enhanced the solubility, facilitating the single-step purification of lengthy hydrophobic peptides. Most importantly, PEGylation did not compromise cell viability and had little to no effect on the immunogenicity of the peptides. In contrast, the addition of a palmitoyl group to increase immunogenicity led to reduced yield and solubility. Overall, PEGylation proves to be an effective technique for enhancing the solubility and broadening the range of utility of diverse long hydrophobic peptides.
Collapse
Affiliation(s)
- Omar F. Luna
- Department
of Organic Chemistry, University of Barcelona,
and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials,
and Nanomedicine, Barcelona 08028, Spain
| | - Yomkippur V. Perez
- Polypure
AS, Martin Linges vei 25, Fornebu 1364, Norway
- Department
of Chemistry, Bioscience, and Environmental Engineering, University of Stavanger, Stavanger 4036, Norway
| | - Daniele P. Ferrari
- Translational
Molecular Imaging, Max Planck Institute
of Multidisciplinary Sciences, Göttingen 37075, Germany
| | - Sana S. Sayedipour
- Department
of Radiology, Leiden University Medical
Center, Leiden 2333, Netherlands
| | - Miriam Royo
- Department
of Organic Chemistry, University of Barcelona,
and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials,
and Nanomedicine, Barcelona 08028, Spain
- Institute
for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council
for Scientific Research (CSIC), Barcelona 08028, Spain
| | - Gerardo A. Acosta
- Department
of Organic Chemistry, University of Barcelona,
and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials,
and Nanomedicine, Barcelona 08028, Spain
- Institute
for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council
for Scientific Research (CSIC), Barcelona 08028, Spain
| | - Luis J. Cruz
- Department
of Radiology, Leiden University Medical
Center, Leiden 2333, Netherlands
| | - Frauke Alves
- Translational
Molecular Imaging, Max Planck Institute
of Multidisciplinary Sciences, Göttingen 37075, Germany
| | - Erik Agner
- Polypure
AS, Martin Linges vei 25, Fornebu 1364, Norway
| | - Magne O. Sydnes
- Department
of Chemistry, Bioscience, and Environmental Engineering, University of Stavanger, Stavanger 4036, Norway
| | - Fernando Albericio
- Department
of Organic Chemistry, University of Barcelona,
and CIBER-BBN, Networking Centre on Bioengineering, Biomaterials,
and Nanomedicine, Barcelona 08028, Spain
- Peptide
Science Laboratory, School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
| |
Collapse
|
21
|
Ning J, Lu X, Dong J, Xue C, Ou C, Zhang Y, Zhang X, Gao F. Advanced Strategies for Strengthening the Immune Activation Effect of Traditional Antitumor Therapies. ACS Biomater Sci Eng 2024; 10:4701-4715. [PMID: 38959418 DOI: 10.1021/acsbiomaterials.4c00560] [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: 07/05/2024]
Abstract
The utilization of traditional therapies (TTS), such as chemotherapy, reactive oxygen species-based therapy, and thermotherapy, to induce immunogenic cell death (ICD) in tumor cells has emerged as a promising strategy for the activation of the antitumor immune response. However, the limited effectiveness of most TTS in inducing the ICD effect of tumors hinders their applications in combination with immunotherapy. To address this challenge, various intelligent strategies have been proposed to strengthen the immune activation effect of these TTS, and then achieve synergistic antitumor efficacy with immunotherapy. These strategies primarily focus on augmenting the tumor ICD effect or facilitating the antigen (released by the ICD tumor cells) presentation process during TTS, and they are systematically summarized in this review. Finally, the existing bottlenecks and prospects of TTS in the application of tumor immune regulation are also discussed.
Collapse
Affiliation(s)
- Jingyi Ning
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Xinxin Lu
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Jianhui Dong
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Chun Xue
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Changjin Ou
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Yizhou Zhang
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Xianzheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Fan Gao
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| |
Collapse
|
22
|
Gong N, Alameh MG, El-Mayta R, Xue L, Weissman D, Mitchell MJ. Enhancing in situ cancer vaccines using delivery technologies. Nat Rev Drug Discov 2024; 23:607-625. [PMID: 38951662 DOI: 10.1038/s41573-024-00974-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/03/2024]
Abstract
In situ cancer vaccination refers to any approach that exploits tumour antigens available at a tumour site to induce tumour-specific adaptive immune responses. These approaches hold great promise for the treatment of many solid tumours, with numerous candidate drugs under preclinical or clinical evaluation and several products already approved. However, there are challenges in the development of effective in situ cancer vaccines. For example, inadequate release of tumour antigens from tumour cells limits antigen uptake by immune cells; insufficient antigen processing by antigen-presenting cells restricts the generation of antigen-specific T cell responses; and the suppressive immune microenvironment of the tumour leads to exhaustion and death of effector cells. Rationally designed delivery technologies such as lipid nanoparticles, hydrogels, scaffolds and polymeric nanoparticles are uniquely suited to overcome these challenges through the targeted delivery of therapeutics to tumour cells, immune cells or the extracellular matrix. Here, we discuss delivery technologies that have the potential to reduce various clinical barriers for in situ cancer vaccines. We also provide our perspective on this emerging field that lies at the interface of cancer vaccine biology and delivery technologies.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, Center for BioAnalytical Chemistry, Hefei National Research Center for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, George Mason University, Fairfax, VA, USA
| | - Rakan El-Mayta
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Yan W, Cao Y, Yin Q, Li Y. Biomimetic Nucleic Acid Drug Delivery Systems for Relieving Tumor Immunosuppressive Microenvironment. Pharmaceutics 2024; 16:1028. [PMID: 39204373 PMCID: PMC11360391 DOI: 10.3390/pharmaceutics16081028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Immunotherapy combats tumors by enhancing the body's immune surveillance and clearance of tumor cells. Various nucleic acid drugs can be used in immunotherapy, such as DNA expressing cytokines, mRNA tumor vaccines, small interfering RNAs (siRNA) knocking down immunosuppressive molecules, and oligonucleotides that can be used as immune adjuvants. Nucleic acid drugs, which are prone to nuclease degradation in the circulation and find it difficult to enter the target cells, typically necessitate developing appropriate vectors for effective in vivo delivery. Biomimetic drug delivery systems, derived from viruses, bacteria, and cells, can protect the cargos from degradation and clearance, and deliver them to the target cells to ensure safety. Moreover, they can activate the immune system through their endogenous activities and active components, thereby improving the efficacy of antitumor immunotherapeutic nucleic acid drugs. In this review, biomimetic nucleic acid delivery systems for relieving a tumor immunosuppressive microenvironment are introduced. Their immune activation mechanisms, including upregulating the proinflammatory cytokines, serving as tumor vaccines, inhibiting immune checkpoints, and modulating intratumoral immune cells, are elaborated. The advantages and disadvantages, as well as possible directions for their clinical translation, are summarized at last.
Collapse
Affiliation(s)
- Wenlu Yan
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Cao
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qi Yin
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
| |
Collapse
|
24
|
Younis A, Gribben J. Immune Checkpoint Inhibitors: Fundamental Mechanisms, Current Status and Future Directions. IMMUNO 2024; 4:186-210. [DOI: 10.3390/immuno4030013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) are a promising form of immunotherapy that have significantly changed the therapeutic landscape for many advanced cancers. They have shown unique clinical benefit against a broad range of tumour types and a strong overall impact on survival in studied patient populations. However, there are still many limitations holding back this immunotherapy from reaching its full potential as a possible curative option for advanced cancer patients. A great deal of research is being undertaken in the hope of driving advancements in this area, building a better understanding of the mechanisms behind immune checkpoint inhibition and ultimately developing more effective, safer, and wider-reaching agents. Taking into account the current literature on this topic, this review aims to explore in depth the basis of the use of ICIs in the treatment of advanced cancers, evaluate its efficacy and safety, consider its current limitations, and finally reflect on what the future holds for this very promising form of cancer immunotherapy.
Collapse
Affiliation(s)
- Abdullah Younis
- Barts and the London School of Medicine and Dentistry, London E1 2AD, UK
| | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6AU, UK
| |
Collapse
|
25
|
Luo Y, Luo X, Ru Y, Zhou X, Liu D, Huang Q, Linghu M, Wu Y, Lv Z, Chen M, Ma Y, Huang Y, Wang J. Copper(II)-Based Nano-Regulator Correlates Cuproptosis Burst and Sequential Immunogenic Cell Death for Synergistic Cancer Immunotherapy. Biomater Res 2024; 28:0039. [PMID: 38938647 PMCID: PMC11208873 DOI: 10.34133/bmr.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/08/2024] [Indexed: 06/29/2024] Open
Abstract
Immunogenic cell death (ICD) of tumor cells serves as a crucial initial signal in the activation of anti-tumor immune responses, holding marked promise in the field of tumor immunotherapy. However, low immunogenicity tumors pose challenges in achieving complete induction of ICD, thereby limiting the response rates of immunotherapy in clinical patients. The emergence of cuproptosis as a new form of regulated cell death has presented a promising strategy for enhanced immunotherapy of low immunogenic tumors. To trigger cuproptosis, copper-ionophore elesclomol (ES) had to be employed for the copper-transporting-mediated process. Herein, we proposed a copper(II)-based metal-organic framework nanoplatform (Cu-MOF) to facilitate a cooperative delivery of encapsulated ES and copper (ES-Cu-MOF) to induce cuproptosis burst and enhance ICD of fibrosarcoma. Our results showed that the ES-Cu-MOF nano-regulator could effectively release Cu2+ and ES in response to the intracellular environment, resulting in elevated mitochondrial ROS generation and initiated cuproptosis of tumor cells. Furthermore, sequential ICDs were significantly triggered via the ES-Cu-MOF nano-regulator to activate the anti-tumor immune response. The results of tumor inhibition experiment indicated that the nano-regulator of ES-Cu-MOF obviously accumulated in the tumor site, inducing ICD for dendritic cell activation. This enabled an increased infiltration of cytotoxic CD8+ T cells and consequently enhanced antitumor immune responses for successfully suppressing fibrosarcoma growth. Thus, the copper(II)-based metal-organic framework nano-regulator offered a promising approach for inducing cuproptosis and cuproptosis-stimulated ICD for cancer immunotherapy.
Collapse
Affiliation(s)
- Yingli Luo
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
- Affiliated Hospital of Jiangnan University,
Jiangnan University, Wuxi, Jiangsu 214062, PR China
| | - Xianyu Luo
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Yi Ru
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Xinru Zhou
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Didi Liu
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Qian Huang
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Maoyuan Linghu
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Yuhang Wu
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Zicheng Lv
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Meimei Chen
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Yinchu Ma
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
- Affiliated Hospital of Jiangnan University,
Jiangnan University, Wuxi, Jiangsu 214062, PR China
| | - Yi Huang
- Wuxi School of Medicine,
Jiangnan University, Wuxi, Jiangsu 214122, PR China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| | - Jilong Wang
- Joint Centre of Translational Medicine, Wenzhou Institute,
University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, PR China
| |
Collapse
|
26
|
Nikolouzakis TK, Chrysos E, Docea AO, Fragkiadaki P, Souglakos J, Tsiaoussis J, Tsatsakis A. Current and Future Trends of Colorectal Cancer Treatment: Exploring Advances in Immunotherapy. Cancers (Basel) 2024; 16:1995. [PMID: 38893120 PMCID: PMC11171065 DOI: 10.3390/cancers16111995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer of the colon and rectum (CRC) has been identified among the three most prevalent types of cancer and cancer-related deaths for both sexes. Even though significant progress in surgical and chemotherapeutic techniques has markedly improved disease-free and overall survival rates in contrast to those three decades ago, recent years have seen a stagnation in these improvements. This underscores the need for new therapies aiming to augment patient outcomes. A number of emerging strategies, such as immune checkpoint inhibitors (ICIs) and adoptive cell therapy (ACT), have exhibited promising outcomes not only in preclinical but also in clinical settings. Additionally, a thorough appreciation of the underlying biology has expanded the scope of research into potential therapeutic interventions. For instance, the pivotal role of altered telomere length in early CRC carcinogenesis, leading to chromosomal instability and telomere dysfunction, presents a promising avenue for future treatments. Thus, this review explores the advancements in CRC immunotherapy and telomere-targeted therapies, examining potential synergies and how these novel treatment modalities intersect to potentially enhance each other's efficacy, paving the way for promising future therapeutic advancements.
Collapse
Affiliation(s)
| | - Emmanuel Chrysos
- Department of General Surgery, University General Hospital of Heraklion, 71110 Heraklion, Greece; (T.K.N.); (E.C.)
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Persefoni Fragkiadaki
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece; (P.F.); (A.T.)
| | - John Souglakos
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece;
| | - John Tsiaoussis
- Department of Anatomy, Medical School, University of Crete, 70013 Heraklion, Greece;
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece; (P.F.); (A.T.)
| |
Collapse
|
27
|
Parveen M, Karaosmanoglu B, Sucularli C, Uner A, Taskiran EZ, Esendagli G. Acquired immune resistance is associated with interferon signature and modulation of KLF6/c-MYB transcription factors in myeloid leukemia. Eur J Immunol 2024; 54:e2350717. [PMID: 38462943 DOI: 10.1002/eji.202350717] [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: 08/16/2023] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 03/12/2024]
Abstract
Resistance to immunity is associated with the selection of cancer cells with superior capacities to survive inflammatory reactions. Here, we tailored an ex vivo immune selection model for acute myeloid leukemia (AML) and isolated the residual subpopulations as "immune-experienced" AML (ieAML) cells. We confirmed that upon surviving the immune reactions, the malignant blasts frequently decelerated proliferation, displayed features of myeloid differentiation and activation, and lost immunogenicity. Transcriptomic analyses revealed a limited number of commonly altered pathways and differentially expressed genes in all ieAML cells derived from distinct parental cell lines. Molecular signatures predominantly associated with interferon and inflammatory cytokine signaling were enriched in the AML cells resisting the T-cell-mediated immune reactions. Moreover, the expression and nuclear localization of the transcription factors c-MYB and KLF6 were noted as the putative markers for immune resistance and identified in subpopulations of AML blasts in the patients' bone marrow aspirates. The immune modulatory capacities of ieAML cells lasted for a restricted period when the immune selection pressure was omitted. In conclusion, myeloid leukemia cells harbor subpopulations that can adapt to the harsh conditions established by immune reactions, and a previous "immune experience" is marked with IFN signature and may pave the way for susceptibility to immune intervention therapies.
Collapse
Affiliation(s)
- Mubaida Parveen
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Türkiye
| | - Beren Karaosmanoglu
- Department of Medical Genetics, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Ceren Sucularli
- Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara, Türkiye
| | - Aysegul Uner
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Ekim Z Taskiran
- Department of Medical Genetics, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Gunes Esendagli
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Türkiye
| |
Collapse
|
28
|
De Velasco MA, Kura Y, Fujita K, Uemura H. Moving toward improved immune checkpoint immunotherapy for advanced prostate cancer. Int J Urol 2024; 31:307-324. [PMID: 38167824 DOI: 10.1111/iju.15378] [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: 08/17/2023] [Accepted: 12/10/2023] [Indexed: 01/05/2024]
Abstract
Human prostate cancer is a heterogenous malignancy that responds poorly to immunotherapy targeting immune checkpoints. The immunosuppressive tumor microenvironment that is typical of human prostate cancer has been the main obstacle to these treatments. The effectiveness of these therapies is also hindered by acquired resistance, leading to slow progress in prostate cancer immunotherapy. Results from the highly anticipated late-stage clinical trials of PD-1/PD-L1 immune checkpoint blockade in patients with advanced prostate cancer have highlighted some of the obstacles to immunotherapy. Despite the setbacks, there is much that has been learned about the mechanisms that drive resistance, and new strategies are being developed and tested. Here, we review the status of immune checkpoint blockade and the immunosuppressive tumor microenvironment and discuss factors contributing to innate and adaptive resistance to immune checkpoint blockade within the context of prostate cancer. We then examine current strategies aiming to overcome these challenges as well as prospects.
Collapse
Affiliation(s)
- Marco A De Velasco
- Department of Genome Biology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Yurie Kura
- Department of Genome Biology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Kazutoshi Fujita
- Department of Urology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kindai University Faculty of Medicine, Osakasayama, Japan
| |
Collapse
|
29
|
Fu W, Li X, Li Y, Luo R, Ou C, Huang D, Liang X, You Y, Wu Q, Gong C. A programmable releasing versatile hydrogel platform boosts systemic immune responses via sculpting tumor immunogenicity and reversing tolerogenic dendritic cells. Biomaterials 2024; 305:122444. [PMID: 38142471 DOI: 10.1016/j.biomaterials.2023.122444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/28/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Immunogenicity improvement is a valuable strategy for tumor immunotherapy. However, immunosuppressive factors bestow tolerogenic phenotype on tumor-infiltrating DCs, which exhibit weak antigen presentation and strong anti-inflammatory cytokines secretion abilities, limiting the effectiveness of tumor immunotherapy even if the tumor has adequate immunogenicity. Herein, we designed a programmable releasing versatile hydrogel platform (PIVOT) to sculpt tumor immunogenicity, increase intratumoral DCs and cDC1s abundance, and reverse the tolerogenic phenotype of DCs, thus promoting their maturation for boosting innate and adaptive immune responses. Responsive to tumoral reactive oxygen species (ROS), the hydrogel splits and promotes the activation of DCs and macrophages. Then, oxaliplatin is first released from PIVOT to sculpt tumor immunogenicity by inducing immunogenic cell death (ICD) and causing tumoral DNA fragments exposure simultaneously. Subsequently, the impaired DNA fragments bind to high mobility group protein 1 (HMGB1) forming the DNA-HMGB1 complex. Moreover, exogenous FMS-like tyrosine kinase 3 ligand (Flt-3L) recruits masses of DCs, especially cDC1s, which will endocytose the complex benefiting from TIM-3 blockade (αTIM3) that can reverse tolerogenic DCs. Finally, the endocytosis activates the cGAS-STING pathway of cDC1s, which promotes the secretion of type I IFN that triggers innate immune responses, and CXCL9 which recruits CD8+ effector T cells to initiate the following adaptive immune response against tumor progress. PIVOT achieves nearly 90 % tumor growth inhibition and induces systemic antitumor immune responses. In conclusion, this study focuses on ICD-mediated tumor immunogenicity sculpture and nucleic acid endocytosis-involved tolerogenic DCs reversal, providing a novel paradigm for enhancing DCs-based antitumor immune responses.
Collapse
Affiliation(s)
- Wangxian Fu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinchao Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yingjie Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunqing Ou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dongxue Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiuqi Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanjie You
- Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
30
|
diZerega GS, Maulhardt HA, Verco SJ, Marin AM, Baltezor MJ, Mauro SA, Iacobucci MA. Intratumoral Injection of Large Surface Area Microparticle Taxanes in Carcinomas Increases Immune Effector Cell Concentrations, Checkpoint Expression, and Synergy with Checkpoint Inhibitors: A Review of Preclinical and Clinical Studies. Oncol Ther 2024; 12:31-55. [PMID: 38289576 PMCID: PMC10881942 DOI: 10.1007/s40487-024-00261-y] [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: 11/03/2023] [Accepted: 01/04/2024] [Indexed: 02/23/2024] Open
Abstract
This review summarizes development of large surface area microparticle paclitaxel (LSAM-PTX) and docetaxel (LSAM-DTX) for local treatment of primary carcinomas with emphasis on immunomodulation. Intratumoral (IT) delivery of LSAM-PTX and LSAM-DTX provides continuous, therapeutic drug levels for several weeks. Preclinical studies and clinical trials reported a reduction in tumor volume (TV) and immunomodulation in primary tumor and peripheral blood with increases in innate and adaptive immune cells and decreases in suppressor cells. Increased levels of checkpoint expression of immune cells occurred in clinical trials of high-risk non-muscle-invasive bladder cancer (LSAM-DTX) and unresectable localized pancreatic cancer (LSAM-PTX). TV reduction and increases in immune effector cells occurred following IT LSAM-DTX and IT LSAM-PTX together with anti-mCTLA-4 and anti-mPD-1, respectively. Synergistic benefits from combinatorial therapy in a 4T1-Luc breast cancer model included reduction of metastasis with IT LSAM-DTX + anti-mCTLA-4. IT LSAM-PTX and LSAM-DTX are tumoricidal, immune enhancing, and may improve solid tumor response to immune checkpoint inhibitors without additional systemic toxicity.
Collapse
Affiliation(s)
- Gere S diZerega
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA.
- NanOlogy, LLC., 3909 Hulen Street, Fort Worth, TX, 76107, USA.
| | - Holly A Maulhardt
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA
| | - Shelagh J Verco
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA
| | - Alyson M Marin
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA
| | | | - Samantha A Mauro
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA
| | | |
Collapse
|
31
|
Feng S, Zhang Y, Zhu H, Jian Z, Zeng Z, Ye Y, Li Y, Smerin D, Zhang X, Zou N, Gu L, Xiong X. Cuproptosis facilitates immune activation but promotes immune escape, and a machine learning-based cuproptosis-related signature is identified for predicting prognosis and immunotherapy response of gliomas. CNS Neurosci Ther 2024; 30:e14380. [PMID: 37515314 PMCID: PMC10848101 DOI: 10.1111/cns.14380] [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: 03/28/2023] [Revised: 06/27/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
AIMS Cell death, except for cuproptosis, in gliomas has been extensively studied, providing novel targets for immunotherapy by reshaping the tumor immune microenvironment through multiple mechanisms. This study aimed to explore the effect of cuproptosis on the immune microenvironment and its predictive power in prognosis and immunotherapy response. METHODS Eight glioma cohorts were included in this study. We employed the unsupervised clustering algorithm to identify novel cuproptosis clusters and described their immune microenvironmental characteristics, mutation landscape, and altered signaling pathways. We verified the correlation among FDX1, SLC31A1, and macrophage infiltration in 56 glioma tissues. Next, based on multicenter cohorts and 10 machine learning algorithms, we constructed an artificial intelligence-driven cuproptosis-related signature named CuproScore. RESULTS Our findings suggested that glioma patients with high levels of cuproptosis had a worse prognosis owing to immunosuppression caused by unique immune escape mechanisms. Meanwhile, we experimentally validated the positive association between cuproptosis and macrophages and its tumor-promoting mechanism in vitro. Furthermore, our CuproScore exhibited powerful and robust prognostic predictive ability. It was also capable of predicting response to immunotherapy and chemotherapy drug sensitivity. CONCLUSIONS Cuproptosis facilitates immune activation but promotes immune escape. The CuproScore could predict prognosis and immunotherapy response in gliomas.
Collapse
Affiliation(s)
- Shi Feng
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yonggang Zhang
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hua Zhu
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhihong Jian
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhi Zeng
- Department of PathologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yingze Ye
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yina Li
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Daniel Smerin
- Department of NeurosurgeryUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Xu Zhang
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Ning Zou
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lijuan Gu
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xiaoxing Xiong
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
32
|
Lv Y, Niu L, Li Q, Shao W, Yan X, Li Y, Yue Y, Chen H. Identification of an immune-related eRNA prognostic signature for clear cell renal cell carcinoma. Aging (Albany NY) 2024; 16:2232-2248. [PMID: 38289619 PMCID: PMC10911372 DOI: 10.18632/aging.205479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/01/2023] [Indexed: 02/22/2024]
Abstract
BACKGROUND Immune-related enhancer RNAs (eRNAs) have garnered significant attention in cancer metabolism research, yet their specific roles in ccRCC have remained elusive. METHODS We retrieved eRNA expression profiles from TCGA database and identified immune-related eRNAs (IREs) by assessing their co-expression with immune genes. Utilizing consensus clustering, we organized these IREs into two distinct clusters. The construction of an IREs signature was accomplished through the LASSO and multivariate Cox analysis. Furthermore, we performed Cell Counting Kit-8 and clonogenic assays to assess changes in the proliferative capacity of Caki-1 and 769-P cells. RESULTS The existence of two clusters of immune-related eRNAs in ccRCC, each with distinctive prognostic and immunological attributes. Cluster B exhibited immunosuppressive properties and displayed a positive correlation with immunosuppressive cells. Functional enrichment analysis unveiled their involvement in several tumor-promoting pathways, metabolic pathways and immune pathways. The IREs signature demonstrated its potential to accurately predict patient immune and prognostic characteristics. AC003092.1, an eRNA strongly associated with patient survival, emerged as a potential oncogene significantly linked to adverse prognosis and the presence of immunosuppressive cells and checkpoints in ccRCC patients. Notably, AC003092.1 displayed marked upregulation in ccRCC tissues and cell lines, and its knockdown substantially inhibited the proliferation of Caki-1 and 769-P cells. CONCLUSION We established a robust predictive model that played a vital role in determining the prognosis, clinicopathological characteristics and immune cell infiltration patterns of ccRCC patients. IRE, particularly AC003092.1, which was strongly associated with survival, hold promise as novel immunotherapeutic targets for ccRCC.
Collapse
Affiliation(s)
- Yang Lv
- Department of Urology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215228, China
| | - Lili Niu
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116021, China
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Qiang Li
- Department of Urology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215228, China
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenchuan Shao
- Department of Urology, The State Key Lab of Reproductive, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xinghan Yan
- Department of Urology, The State Key Lab of Reproductive, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yang Li
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Yulin Yue
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Hongqi Chen
- Department of Urology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 215228, China
| |
Collapse
|
33
|
Tavira B, Iscar T, Manso L, Santaballa A, Gil-Martin M, García García Y, Romeo M, Iglesias M, de Juan Ferré A, Barretina-Ginesta MP, Manzano A, Gaba L, Rubio MJ, de Andrea CE, González-Martín A. Analysis of Tumor Microenvironment Changes after Neoadjuvant Chemotherapy with or without Bevacizumab in Advanced Ovarian Cancer (GEICO-89T/MINOVA Study). Clin Cancer Res 2024; 30:176-186. [PMID: 37527007 PMCID: PMC10767307 DOI: 10.1158/1078-0432.ccr-23-0771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/08/2023] [Accepted: 07/27/2023] [Indexed: 08/03/2023]
Abstract
PURPOSE The aim of our study was to elucidate the impact of bevacizumab added to neoadjuvant chemotherapy (NACT) on the tumor immune microenvironment and correlate the changes with the clinical outcome of the patients. EXPERIMENTAL DESIGN IHC and multiplex immunofluorescence for lymphoid and myeloid lineage markers were performed in matched tumor samples from 23 patients with ovarian cancer enrolled in GEICO 1205/NOVA clinical study before NACT and at the time of interval cytoreductive surgery. RESULTS Our results showed that the addition of bevacizumab to NACT plays a role mainly on lymphoid populations at the stromal compartment, detecting a significant decrease of CD4+ T cells, an increase of CD8+ T cells, and an upregulation in effector/regulatory cell ratio (CD8+/CD4+FOXP3+). None of the changes observed were detected in the intra-epithelial site in any arm (NACT or NACT-bevacizumab). No differences were found in myeloid lineage (macrophage-like). The percentage of Treg populations and effector/regulatory cell ratio in the stroma were the only two variables significantly associated with progression-free survival (PFS). CONCLUSIONS The addition of bevacizumab to NACT did not have an impact on PFS in the GEICO 1205 study. However, at the cellular level, changes in CD4+, CD8+ lymphocyte populations, and CD8+/CD4+FOXP3 ratio have been detected only at the stromal site. On the basis of our results, we hypothesize about the existence of mechanisms of resistance that could prevent the trafficking of T-effector cells into the epithelial component of the tumor as a potential explanation for the lack of efficacy of ICI in the first-line treatment of advanced epithelial ovarian cancer. See related commentary by Soberanis Pina and Oza, p. 12.
Collapse
Affiliation(s)
- Beatriz Tavira
- Laboratory of Translational Oncology, Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Teresa Iscar
- Department of Pathology, Cancer Center Clínica Universidad de Navarra, Madrid, Spain
| | - Luis Manso
- Department of Medical Oncology, Hospital 12 de Octubre, Madrid, Spain
| | - Ana Santaballa
- Department of Medical Oncology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Marta Gil-Martin
- Department of Medical Oncology, Institut Català d'Oncologia L'Hospitalet, Hospitalet de Llobregat, Spain
| | - Yolanda García García
- Department of Medical Oncology, Parc Taulí Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Margarita Romeo
- Department of Medical Oncology, Institut Català d'Oncologia Badalona, Badalona, Spain
| | - Maria Iglesias
- Department of Medical Oncology, Hospital Son Llátzer, Palma de Mallorca, Spain
| | - Ana de Juan Ferré
- Department of Medical Oncology, Hospital Marqués de Valdecilla, Santander, Spain
| | | | - Aranzazu Manzano
- Department of Medical Oncology, Hospital Clínico San Carlos, Madrid, Spain
| | - Lydia Gaba
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - María Jesús Rubio
- Department of Medical Oncology, Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Carlos E. de Andrea
- Department of Pathology, Cancer Center Clínica Universidad de Navarra, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Antonio González-Martín
- Laboratory of Translational Oncology, Program in Solid Tumors, Cima-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Department of Medical Oncology, Cancer Center Clínica Universidad de Navarra, Madrid, Spain
| |
Collapse
|
34
|
Mao M, Jiang F, Han R, Xiang Y. Identification of the prognostic immune subtype in copy-number high endometrial cancer. J Gynecol Oncol 2024; 35:e8. [PMID: 37857563 PMCID: PMC10792215 DOI: 10.3802/jgo.2024.35.e8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 10/21/2023] Open
Abstract
OBJECTIVE The TCGA molecular subtype of endometrial cancer (EC) is widely applied, among which the copy-number high (CNH) subtype has the poorest prognosis. However, the heterogeneity of this subtype remains elusive. In this study, we aimed to identify heterogeneous immune subtypes in CNH EC and explore their prognostic significance. METHODS We collected 60 CNH EC cases in the TCGA database and performed unsupervised cluster analysis based on the enrichment scores of immune-related gene signatures to identify immune subtypes. We described their immune characteristics and prognoses and conducted differential gene analysis and lasso regression to identify a prognostic biomarker, GZMM. For experimental validation, we performed immunohistochemical staining of GZMM in 39 p53-positive EC surgical samples. RESULTS We defined two immune subtypes, immune-hot (IH) and immune-cold (IC), which differed in immune cell infiltration, cytokine and chemokine expression and prognosis. The IH subtype has significantly stronger immune activation than the IC subtype, showing a significant infiltration of immune effector cells and high expression of relevant chemokines, with better prognosis. Moreover, the immunohistochemical staining of GZMM in a cohort of 39 p53-positive EC surgical samples confirmed GZMM as a unique prognostic biomarker, with high expression in both tumor cells and lymphocytes predicting a better prognosis. CONCLUSION Our study revealed heterogeneous immune subtypes in CNH EC and identified GZMM as a prognostic biomarker. The stratified classification strategy combining molecular and immune subtypes provides valuable insights for future clinical practice.
Collapse
Affiliation(s)
- Mingyi Mao
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Fang Jiang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China.
| | - Ruiqin Han
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Xiang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| |
Collapse
|
35
|
Cho Y, Lee J, Han B, Yoon SE, Kim SJ, Kim WS, Cho J. Tumor-infiltrating T lymphocytes evaluated using digital image analysis predict the prognosis of patients with diffuse large B-cell lymphoma. J Pathol Transl Med 2024; 58:12-21. [PMID: 38229430 DOI: 10.4132/jptm.2023.11.02] [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: 10/10/2023] [Accepted: 11/01/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND The implication of the presence of tumor-infiltrating T lymphocytes (TIL-T) in diffuse large B-cell lymphoma (DLBCL) is yet to be elucidated. We aimed to investigate the effect of TIL-T levels on the prognosis of patients with DLBCL. METHODS Ninety-six patients with DLBCL were enrolled in the study. The TIL-T ratio was measured using QuPath, a digital pathology software package. The TIL-T ratio was investigated in three foci (highest, intermediate, and lowest) for each case, resulting in TIL-T-Max, TIL-T-Intermediate, and TIL-T-Min. The relationship between the TIL-T ratios and prognosis was investigated. RESULTS When 19% was used as the cutoff value for TIL-T-Max, 72 (75.0%) and 24 (25.0%) patients had high and low TIL-T-Max, respectively. A high TIL-T-Max was significantly associated with lower serum lactate dehydrogenase levels (p < .001), with patient group who achieved complete remission after RCHOP therapy (p < .001), and a low-risk revised International Prognostic Index score (p < .001). Univariate analysis showed that patients with a low TIL-T-Max had a significantly worse prognosis in overall survival compared to those with a high TIL-T-Max (p < .001); this difference remained significant in a multivariate analysis with Cox proportional hazards (hazard ratio, 7.55; 95% confidence interval, 2.54 to 22.42; p < .001). CONCLUSIONS Patients with DLBCL with a high TIL-T-Max showed significantly better prognosis than those with a low TIL-T-Max, and the TIL-T-Max was an independent indicator of overall survival. These results suggest that evaluating TIL-T ratios using a digital pathology system is useful in predicting the prognosis of patients with DLBCL.
Collapse
Affiliation(s)
- Yunjoo Cho
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jiyeon Lee
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Pathology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Bogyeong Han
- Department of Pathology, Seoul National University, Seoul National College of Medicine, Seoul, Korea
| | - Sang Eun Yoon
- Division of Hematology and Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Jin Kim
- Division of Hematology and Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Seog Kim
- Division of Hematology and Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Junhun Cho
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
36
|
Janssens S, Rennen S, Agostinis P. Decoding immunogenic cell death from a dendritic cell perspective. Immunol Rev 2024; 321:350-370. [PMID: 38093416 DOI: 10.1111/imr.13301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Dendritic cells (DCs) are myeloid cells bridging the innate and adaptive immune system. By cross-presenting tumor-associated antigens (TAAs) liberated upon spontaneous or therapy-induced tumor cell death to T cells, DCs occupy a pivotal position in the cancer immunity cycle. Over the last decades, the mechanisms linking cancer cell death to DC maturation, have been the focus of intense research. Growing evidence supports the concept that the mere transfer of TAAs during the process of cell death is insufficient to drive immunogenic DC maturation unless this process is coupled with the release of immunomodulatory signals by dying cancer cells. Malignant cells succumbing to a regulated cell death variant called immunogenic cell death (ICD), foster a proficient interface with DCs, enabling their immunogenic maturation and engagement of adaptive immunity against cancer. This property relies on the ability of ICD to exhibit pathogen-mimicry hallmarks and orchestrate the emission of a spectrum of constitutively present or de novo-induced danger signals, collectively known as damage-associated molecular patterns (DAMPs). In this review, we discuss how DCs perceive and decode danger signals emanating from malignant cells undergoing ICD and provide an outlook of the major signaling and functional consequences of this interaction for DCs and antitumor immunity.
Collapse
Affiliation(s)
- Sophie Janssens
- Laboratory for ER Stress and Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sofie Rennen
- Laboratory for ER Stress and Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Patrizia Agostinis
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
37
|
Gehl V, O'Rourke CJ, Andersen JB. Immunogenomics of cholangiocarcinoma. Hepatology 2023:01515467-990000000-00649. [PMID: 37972940 DOI: 10.1097/hep.0000000000000688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023]
Abstract
The development of cholangiocarcinoma spans years, if not decades, during which the immune system becomes corrupted and permissive to primary tumor development and metastasis. This involves subversion of local immunity at tumor sites, as well as systemic immunity and the wider host response. While immune dysfunction is a hallmark of all cholangiocarcinoma, the specific steps of the cancer-immunity cycle that are perturbed differ between patients. Heterogeneous immune functionality impacts the evolutionary development, pathobiological behavior, and therapeutic response of these tumors. Integrative genomic analyses of thousands of primary tumors have supported a biological rationale for immune-based stratification of patients, encompassing immune cell composition and functionality. However, discerning immune alterations responsible for promoting tumor initiation, maintenance, and progression from those present as bystander events remains challenging. Functionally uncoupling the tumor-promoting or tumor-suppressing roles of immune profiles will be critical for identifying new immunomodulatory treatment strategies and associated biomarkers for patient stratification. This review will discuss the immunogenomics of cholangiocarcinoma, including the impact of genomic alterations on immune functionality, subversion of the cancer-immunity cycle, as well as clinical implications for existing and novel treatment strategies.
Collapse
Affiliation(s)
- Virag Gehl
- Department of Health and Medical Sciences, Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
38
|
Huang Y, Lin A, Gu T, Hou S, Yao J, Luo P, Zhang J. CACNA1C mutation as a prognosis predictor of immune checkpoint inhibitor in skin cutaneous melanoma. Immunotherapy 2023; 15:1275-1291. [PMID: 37584225 DOI: 10.2217/imt-2022-0175] [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: 08/17/2023] Open
Abstract
Aims: There is an urgent need for appropriate biomarkers that can precisely and reliably predict immunotherapy efficacy, as immunotherapy responses can differ in skin cutaneous melanoma (SKCM) patients. Methods: In this study, univariate regression models and survival analysis were used to examine the link between calcium voltage-gated channel subunit alpha 1C (CACNA1C) mutation status and immunotherapy outcome in SKCM patients receiving immunotherapy. Mutational landscape, immunogenicity, tumor microenvironment and pathway-enrichment analyses were also performed. Results: The CACNA1C mutation group had a better prognosis, higher immunogenicity, lower endothelial cell infiltration, significant enrichment of antitumor immune response pathways and significant downregulation of protumor pathways. Conclusion: CACNA1C mutation status is anticipated to be a biomarker for predicting melanoma immunotherapy effectiveness.
Collapse
Affiliation(s)
- Yushan Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Tianqi Gu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuang Hou
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Jiarong Yao
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| |
Collapse
|
39
|
Gibbons E, Minor BMN, Hammes SR. Lymphangioleiomyomatosis: where endocrinology, immunology and tumor biology meet. Endocr Relat Cancer 2023; 30:e230102. [PMID: 37410387 PMCID: PMC10529736 DOI: 10.1530/erc-23-0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
Abstract Lymphangioleiomyomatosis (LAM) is a cystic lung disease found almost exclusively in genetic females and caused by small clusters of smooth muscle cell tumors containing mutations in one of the two tuberous sclerosis genes (TSC1 or TSC2). Significant advances over the past 2-3 decades have allowed researchers and clinicians to more clearly understand the pathophysiology of LAM, and therefore better diagnose and treat patients with this disease. Despite substantial progress, only one proven treatment for LAM is used in practice: mechanistic target of rapamycin complex 1 (mTORC1) inhibition with medications such as sirolimus. While mTORC1 inhibition effectively slows LAM progression in many patients, it is not curative, is not effective in all patients, and can be associated with significant side effects. Furthermore, the presence of established and accurate biomarkers to follow LAM progression is limited. That said, discovering additional diagnostic and treatment options for LAM is paramount. This review will describe recent advances in LAM research, centering on the origin and nature of the LAM cell, the role of estrogen in LAM progression, the significance of melanocytic marker expression in LAM cells, and the potential roles of the microenvironment in promoting LAM tumor growth. By appreciating these processes in more detail, researchers and caregivers may be afforded novel approaches to aid in the treatment of patients with LAM.
Collapse
Affiliation(s)
- Erin Gibbons
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Briaunna M. N. Minor
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Stephen R Hammes
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
40
|
Dai YW, Wang WM, Zhou X. Development of a CD8 + T cell-based molecular classification for predicting prognosis and heterogeneity in triple-negative breast cancer by integrated analysis of single-cell and bulk RNA-sequencing. Heliyon 2023; 9:e19798. [PMID: 37810147 PMCID: PMC10559128 DOI: 10.1016/j.heliyon.2023.e19798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC), although the most intractable subtype, is characterized by abundant immunogenicity, which enhances responsiveness to immunotherapeutic measures. Methods First, we identified CD8+ T cell core genes (TRCG) based on single-cell sequence and traditional transcriptome sequencing and then used this data to develop a first-of-its-kind classification system based on CD8+ T cells in patients with TNBC. Next, TRCG-related patterns were systematically analyzed, and their correlation with genomic features, immune activity (microenvironment associated with immune infiltration), and clinicopathological characteristics were assessed in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC), the Cancer Genome Atlas (TCGA), GSE103091, GSE96058 databases. Additionally, a CD8+ T cell-related prognostic signature (TRPS) was developed to quantify a patient-specific TRCG pattern. What's more, the genes-related TRPS was validated by polymerase chain reaction (PCR) experiment. Results This study, for the first time, distinguished two subsets in patients with TNBC based on the TRCG. The immune microenvironment and prognostic stratification between these have distinct heterogeneity. Furthermore, this study constructed a novel scoring system named TRPS, which we show to be a robust prognostic marker for TNBC that is related to the intensity of immune infiltration and immunotherapy. Moreover, the levels of genes related the TRPS were validated by quantitative Real-Time PCR. Conclusions Consequently, this study unraveled an association between the TRCG and the tumor microenvironment in TNBC. TRPS model represents an effective tool for survival prediction and treatment guidance in TNBC that can also help identify individual variations in TME and stratify patients who are sensitive to anticancer immunotherapy.
Collapse
Affiliation(s)
- Yin-wei Dai
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Wei-ming Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiang Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, China
| |
Collapse
|
41
|
Guterres A, Abrahim M, da Costa Neves PC. The role of immune subtyping in glioma mRNA vaccine development. Immunotherapy 2023; 15:1057-1072. [PMID: 37431617 DOI: 10.2217/imt-2023-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023] Open
Abstract
Studies on the development of mRNA vaccines for central nervous system tumors have used gene expression profiles, clinical data and RNA sequencing from sources such as The Cancer Genome Atlas and Chinese Glioma Genome Atlas to identify effective antigens. These studies revealed several immune subtypes of glioma, each one linked to unique prognoses and genetic/immune-modulatory changes. Potential antigens include ARPC1B, BRCA2, COL6A1, ITGB3, IDH1, LILRB2, TP53 and KDR, among others. Patients with immune-active and immune-suppressive phenotypes were found to respond better to mRNA vaccines. While these findings indicate the potential of mRNA vaccines in cancer therapy, further research is required to optimize administration and adjuvant selection, and precisely identify target antigens.
Collapse
Affiliation(s)
- Alexandro Guterres
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Vice-Diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ, 21040-360, Brazil
| | - Mayla Abrahim
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Vice-Diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ, 21040-360, Brazil
| | - Patrícia Cristina da Costa Neves
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos, Vice-Diretoria de Desenvolvimento Tecnológico, Bio-Manguinhos, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ, 21040-360, Brazil
| |
Collapse
|
42
|
Yu J, Li M, Ren B, Cheng L, Wang X, Ma Z, Yong WP, Chen X, Wang L, Goh BC. Unleashing the efficacy of immune checkpoint inhibitors for advanced hepatocellular carcinoma: factors, strategies, and ongoing trials. Front Pharmacol 2023; 14:1261575. [PMID: 37719852 PMCID: PMC10501787 DOI: 10.3389/fphar.2023.1261575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/18/2023] [Indexed: 09/19/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent primary liver cancer, representing approximately 85% of cases. The diagnosis is often made in the middle and late stages, necessitating systemic treatment as the primary therapeutic option. Despite sorafenib being the established standard of care for advanced HCC in the past decade, the efficacy of systemic therapy remains unsatisfactory, highlighting the need for novel treatment modalities. Recent breakthroughs in immunotherapy have shown promise in HCC treatment, particularly with immune checkpoint inhibitors (ICIs). However, the response rate to ICIs is currently limited to approximately 15%-20% of HCC patients. Recently, ICIs demonstrated greater efficacy in "hot" tumors, highlighting the urgency to devise more effective approaches to transform "cold" tumors into "hot" tumors, thereby enhancing the therapeutic potential of ICIs. This review presented an updated summary of the factors influencing the effectiveness of immunotherapy in HCC treatment, identified potential combination therapies that may improve patient response rates to ICIs, and offered an overview of ongoing clinical trials focusing on ICI-based combination therapy.
Collapse
Affiliation(s)
- Jiahui Yu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Mengnan Li
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Boxu Ren
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Le Cheng
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Xiaoxiao Wang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Wei Peng Yong
- Department of Haematology–Oncology, National University Cancer Institute, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaoguang Chen
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Boon Cher Goh
- Department of Haematology–Oncology, National University Cancer Institute, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| |
Collapse
|
43
|
Bandopadhyay S, Patranabis S. Mechanisms of HIF-driven immunosuppression in tumour microenvironment. J Egypt Natl Canc Inst 2023; 35:27. [PMID: 37646847 DOI: 10.1186/s43046-023-00186-z] [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: 06/19/2023] [Accepted: 08/12/2023] [Indexed: 09/01/2023] Open
Abstract
Hypoxia arises due to insufficient oxygen delivery to rapidly proliferating tumour cells that outpace the available blood supply. It is a characteristic feature of most solid tumour microenvironments and plays a critical role in regulating anti-tumour immunity, enhancing tumoral heterogeneity, and promoting therapeutic resistance and poor clinical outcomes. Hypoxia-inducible factors (HIFs) are the major hypoxia-responsive transcription factors that are activated under low oxygenation conditions and have been identified to drive multifunctional roles in tumour immune evasion. The HIF signalling network serves as an attractive target for targeted therapeutic approaches. This review aims to provide a comprehensive overview of the most crucial mechanisms by which HIF controls the expression of immunosuppressive molecules and immune checkpoints, disrupts cancer immunogenicity, and induces immunotherapeutic resistance.
Collapse
Affiliation(s)
| | - Somi Patranabis
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, India.
| |
Collapse
|
44
|
Kan LLY, Chan BCL, Leung PC, Wong CK. Natural-Product-Derived Adjunctive Treatments to Conventional Therapy and Their Immunoregulatory Activities in Triple-Negative Breast Cancer. Molecules 2023; 28:5804. [PMID: 37570775 PMCID: PMC10421415 DOI: 10.3390/molecules28155804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an invasive and persistent subtype of breast cancer that is likely to be resistant to conventional treatments. The rise in immunotherapy has created new modalities to treat cancer, but due to high costs and unreliable efficacy, adjunctive and complementary treatments have sparked interest in enhancing the efficacy of currently available treatments. Natural products, which are bioactive compounds derived from natural sources, have historically been used to treat or ameliorate inflammatory diseases and symptoms. As TNBC patients have shown little to no response to immunotherapy, the potential of natural products as candidates for adjuvant immunotherapy is being explored, as well as their immunomodulatory effects on cancer. Due to the complexity of TNBC and the ever-changing tumor microenvironment, there are challenges in determining the feasibility of using natural products to enhance the efficacy or counteract the toxicity of conventional treatments. In view of technological advances in molecular docking, pharmaceutical networking, and new drug delivery systems, natural products show promise as potential candidates in adjunctive therapy. In this article, we summarize the mechanisms of action of selected natural-product-based bioactive compounds and analyze their roles and applications in combination treatments and immune regulation.
Collapse
Affiliation(s)
- Lea Ling-Yu Kan
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; (L.L.-Y.K.); (B.C.-L.C.); (P.-C.L.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ben Chung-Lap Chan
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; (L.L.-Y.K.); (B.C.-L.C.); (P.-C.L.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ping-Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; (L.L.-Y.K.); (B.C.-L.C.); (P.-C.L.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Chun-Kwok Wong
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; (L.L.-Y.K.); (B.C.-L.C.); (P.-C.L.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China
- Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
45
|
Ning Y, Fang S, Fang J, Lin K, Nie H, Xiong P, Qiu P, Zhao Q, Wang H, Wang F. Guanylate-binding proteins signature predicts favorable prognosis, immune-hot microenvironment, and immunotherapy response in hepatocellular carcinoma. Cancer Med 2023; 12:17504-17521. [PMID: 37551111 PMCID: PMC10501289 DOI: 10.1002/cam4.6347] [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: 12/02/2022] [Revised: 07/01/2023] [Accepted: 07/06/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND The role of guanylate-binding proteins (GBPs) in various cancers has been elucidated recently. However, our knowledge of the clinical relevance and biological characteristics of GBPs in hepatocellular carcinoma (HCC) remains limited. METHODS A total of 955 HCC patients were enrolled from five independent public HCC cohorts. The role of GBP molecules in HCC was preliminarily investigated, and a GBP family signature, termed GBPs-score, was constructed by principal component analysis to combine the GBP molecule values. We revealed the effects of GBP genes and GBPs-score in HCC via well-established bioinformatics methods and validated GBP1-5 experimentally in a tissue microarray (TMA) cohort. RESULTS GBPs molecules were closely associated with the prognosis of patients with HCC, and a high GBPs-score highly inferred a favorable survival outcome. We also revealed high GBPs-score was related to anti-tumor immunity, the immune-hot tumor microenvironment (TME), and immunotherapy response. Among the GBPs members, GBP1-5 rather than GBP6/7 may be dominant in these fields. The TMA analysis based on immunohistochemistry showed positive correlations between GBP1-5 and the immune-hot TME with abundant infiltration of CD8+ T cells in HCC. CONCLUSIONS Our integrative study revealed the genetic and immunologic characterizations of GBPs in HCC and highlighted their potential values as promising biomarkers for prognosis and immunotherapy.
Collapse
Affiliation(s)
- Yumei Ning
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal DiseasesWuhanChina
| | - Shilin Fang
- Department of Infectious DiseaseZhongnan Hospital of Wuhan University, Hubei AIDS Clinical Training CenterWuhanChina
| | - Jun Fang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Renmin Hospital of Huangmei CountyHuanggangChina
| | - Kun Lin
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal DiseasesWuhanChina
| | - Haihang Nie
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal DiseasesWuhanChina
| | - Peiling Xiong
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal DiseasesWuhanChina
| | - Peishan Qiu
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal DiseasesWuhanChina
| | - Qiu Zhao
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal DiseasesWuhanChina
| | - Haizhou Wang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal DiseasesWuhanChina
| | - Fan Wang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal DiseasesWuhanChina
| |
Collapse
|
46
|
Schunke J, Mailänder V, Landfester K, Fichter M. Delivery of Immunostimulatory Cargos in Nanocarriers Enhances Anti-Tumoral Nanovaccine Efficacy. Int J Mol Sci 2023; 24:12174. [PMID: 37569548 PMCID: PMC10419017 DOI: 10.3390/ijms241512174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Finding a long-term cure for tumor patients still represents a major challenge. Immunotherapies offer promising therapy options, since they are designed to specifically prime the immune system against the tumor and modulate the immunosuppressive tumor microenvironment. Using nucleic-acid-based vaccines or cellular vaccines often does not achieve sufficient activation of the immune system in clinical trials. Additionally, the rapid degradation of drugs and their non-specific uptake into tissues and cells as well as their severe side effects pose a challenge. The encapsulation of immunomodulatory molecules into nanocarriers provides the opportunity of protected cargo transport and targeted uptake by antigen-presenting cells. In addition, different immunomodulatory cargos can be co-delivered, which enables versatile stimulation of the immune system, enhances anti-tumor immune responses and improves the toxicity profile of conventional chemotherapeutic agents.
Collapse
Affiliation(s)
- Jenny Schunke
- Department of Dermatology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Max Planck Insitute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Volker Mailänder
- Department of Dermatology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Max Planck Insitute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | | | - Michael Fichter
- Department of Dermatology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Max Planck Insitute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
47
|
Khodayari S, Khodayari H, Saeedi E, Mahmoodzadeh H, Sadrkhah A, Nayernia K. Single-Cell Transcriptomics for Unlocking Personalized Cancer Immunotherapy: Toward Targeting the Origin of Tumor Development Immunogenicity. Cancers (Basel) 2023; 15:3615. [PMID: 37509276 PMCID: PMC10377122 DOI: 10.3390/cancers15143615] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer immunotherapy is a promising approach for treating malignancies through the activation of anti-tumor immunity. However, the effectiveness and safety of immunotherapy can be limited by tumor complexity and heterogeneity, caused by the diverse molecular and cellular features of tumors and their microenvironments. Undifferentiated tumor cell niches, which we refer to as the "Origin of Tumor Development" (OTD) cellular population, are believed to be the source of these variations and cellular heterogeneity. From our perspective, the existence of distinct features within the OTD is expected to play a significant role in shaping the unique tumor characteristics observed in each patient. Single-cell transcriptomics is a high-resolution and high-throughput technique that provides insights into the genetic signatures of individual tumor cells, revealing mechanisms of tumor development, progression, and immune evasion. In this review, we explain how single-cell transcriptomics can be used to develop personalized cancer immunotherapy by identifying potential biomarkers and targets specific to each patient, such as immune checkpoint and tumor-infiltrating lymphocyte function, for targeting the OTD. Furthermore, in addition to offering a possible workflow, we discuss the future directions of, and perspectives on, single-cell transcriptomics, such as the development of powerful analytical tools and databases, that will aid in unlocking personalized cancer immunotherapy through the targeting of the patient's cellular OTD.
Collapse
Affiliation(s)
- Saeed Khodayari
- International Center for Personalized Medicine (P7MEDICINE), Luise-Rainer-Str. 6-12, 40235 Düsseldorf, Germany
| | - Hamid Khodayari
- International Center for Personalized Medicine (P7MEDICINE), Luise-Rainer-Str. 6-12, 40235 Düsseldorf, Germany
| | - Elnaz Saeedi
- Oxford Clinical Trials Research Unit, Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX3 7LD, UK
| | - Habibollah Mahmoodzadeh
- Breast Disease Research Center, Tehran University of Medical Sciences, Tehran 1819613844, Iran
| | | | - Karim Nayernia
- International Center for Personalized Medicine (P7MEDICINE), Luise-Rainer-Str. 6-12, 40235 Düsseldorf, Germany
| |
Collapse
|
48
|
Peng Z, Li M, Li H, Gao Q. PD-1/PD-L1 immune checkpoint blockade in ovarian cancer: dilemmas and opportunities. Drug Discov Today 2023:103666. [PMID: 37302543 DOI: 10.1016/j.drudis.2023.103666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized treatment in oncology. Antibodies against PD-1/PD-L1 and ICI-based combinations are under clinical investigations in multiple cancers, including ovarian cancer. However, the success of ICIs has not materialized in ovarian cancer, which remains one of the few malignancies where ICIs exhibit modest efficacy as either monotherapy or combination therapy. In this review, we summarize completed and ongoing clinical trials of PD-1/PD-L1 blockade in ovarian cancer, categorize the underlying mechanisms of resistance emergence, and introduce candidate approaches to rewire the tumor microenvironment (TME) to potentiate anti-PD-1/PD-L1 antibodies. Teaser: The intrinsic resistance of ovarian cancer to PD-1/PD-L1 blockade could be overcome by advanced understanding of underlying mechanisms and discoveries of new actionable targets for combinatory treatment.
Collapse
Affiliation(s)
- Zikun Peng
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Li
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huayi Li
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinglei Gao
- Department of Gynaecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; National Clinical Research Centre for Obstetrics and Gynaecology, Cancer Biology Research Centre (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
49
|
Zheng Y, Liu X, Li N, Zhao A, Sun Z, Wang M, Luo J. Radiotherapy combined with immunotherapy could improve the immune infiltration of melanoma in mice and enhance the abscopal effect. Radiat Oncol J 2023; 41:129-139. [PMID: 37403355 DOI: 10.3857/roj.2023.00185] [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: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 07/06/2023] Open
Abstract
PURPOSE To analyze the gene mutation, immune infiltration and tumor growth of primary tumor and distant tumor under different treatment modes. MATERIALS AND METHODS Twenty B16 murine melanoma cells were injected subcutaneously into the of both sides of the thigh, simulating a primary tumor and a secondary tumor impacted by the abscopal effect, respectively. They were divided into blank control group, immunotherapy group, radiotherapy group, and radiotherapy combined immunotherapy group. During this period, tumor volume was measured, and RNA sequencing was performed on tumor samples after the test. R software was used to analyze differentially expressed genes, functional enrichment, and immune infiltration. RESULTS We found that any treatment mode could cause changes in differentially expressed genes, especially the combination treatment. The different therapeutic effects might be caused by gene expression. In addition, the proportions of infiltrating immune cells in the irradiated and abscopal tumors were different. In the combination treatment group, T-cell infiltration in the irradiated site was the most obvious. In the immunotherapy group, CD8+ T-cell infiltration in the abscopal tumor site was obvious, but immunotherapy alone might have a poor prognosis. Whether the irradiated or abscopal tumor was evaluated, radiotherapy combined with anti-programmed cell death protein 1 (anti-PD-1) therapy produced the most obvious tumor control and might have a positive impact on prognosis. CONCLUSION Combination therapy not only improves the immune microenvironment but may also have a positive impact on prognosis.
Collapse
Affiliation(s)
- Yufeng Zheng
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Xue Liu
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Department of Radiotherapy, Dalian Medical University, Dalian, China
| | - Na Li
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Aimei Zhao
- Department of Obstetrics and Gynecology, Liaocheng Dongchangfu District Maternal and Child Health Hospital, Liaocheng, China
| | - Zhiqiang Sun
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Meihua Wang
- Department of Pathology, Changzhou Fourth People's Hospital, Changzhou, China
| | - Judong Luo
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
50
|
Veneroni MV, Festa BM, Costantino A, Spriano G, Mercante G, De Virgilio A, Di Tommaso L. Prognostic Impact of Tumor Immune Microenvironment and Its Predictive Role in Salivary Gland Cancer. Head Neck Pathol 2023; 17:515-527. [PMID: 36723850 PMCID: PMC10293532 DOI: 10.1007/s12105-023-01528-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/09/2023] [Indexed: 02/02/2023]
Abstract
BACKGROUND Recently, many studies have investigated the role of tumor immune microenvironment (TIME) in carcinogenesis, highlighting its relation to both tumor regression and progression. In particular, the "inflammatory system", made of innate and adaptive immune cells, interacts with cancer cells and their surrounding stroma. In this setting, the aim of this review is to summarize the current literature regarding the TIME of major salivary gland carcinomas (MSGCs), with particular attention on the characteristics and prognostic role of tumor infiltrating lymphocytes (TILs), the mechanisms that lead to TILs exhaustion and the important additional immune infiltrating factors that help SGC progression or remission. METHODS A comprehensive literature search was performed concerning published articles on the role of TIME in MSGCs. RESULTS In this work we summarize the advancing knowledge on TIME in SGCs by demonstrating the key prognostic and/or predictive value of specific immune features. CONCLUSION From the analysis of the current 'status of the art' it clearly emerges a need for precise, unambiguous phenotyping of immune cell populations, as well as a more thorough understanding of the frequencies and interactions of multiple immune cell types inside the TIME and their spatial localization (intratumoral vs. stromal).
Collapse
Affiliation(s)
- Maria Vittoria Veneroni
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20090, Pieve Emanuele, MI, Italy
| | - Bianca Maria Festa
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20090, Pieve Emanuele, MI, Italy
- Otorhinolaryngology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, MI, Italy
| | - Andrea Costantino
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20090, Pieve Emanuele, MI, Italy
- Otorhinolaryngology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, MI, Italy
| | - Giuseppe Spriano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20090, Pieve Emanuele, MI, Italy
- Otorhinolaryngology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, MI, Italy
| | - Giuseppe Mercante
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20090, Pieve Emanuele, MI, Italy
- Otorhinolaryngology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, MI, Italy
| | - Armando De Virgilio
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20090, Pieve Emanuele, MI, Italy.
- Otorhinolaryngology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, MI, Italy.
| | - Luca Di Tommaso
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20090, Pieve Emanuele, MI, Italy
- Pathology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, MI, Italy
| |
Collapse
|