1
|
Bhutani K, Vishwakarma S, Yadav P, Yadav MK. The current landscape of aromatase inhibitors for the treatment of estrogen receptor-positive breast carcinoma. J Steroid Biochem Mol Biol 2025; 250:106729. [PMID: 40056742 DOI: 10.1016/j.jsbmb.2025.106729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/10/2025]
Abstract
Estrogen receptor-positive (ER+) breast carcinoma represents a significant portion of breast cancer cases and is characterized by the presence of estrogen receptors that promote tumor growth upon estrogen binding. ER + breast cancer progression involves hormonal influences, interactions within the tumor microenvironment, and genetic mutations that may lead to treatment resistance. Successful therapeutic options include hormonal therapies, particularly aromatase inhibitors (AIs), which aim to block the effects of estrogen or reduce its synthesis. With higher efficacy than tamoxifen, AIs such as anastrozole, letrozole, and exemestane have become widely employed in adjuvant and first-line treatments for advanced breast cancer. AIs function by inhibiting the enzyme aromatase, which converts androgens into estrogens in the peripheral tissues. Because too much estrogen might promote tumor growth, this decrease in estrogen levels is essential for treating ER+ malignancies. To provide a comprehensive overview of AIs in the treatment of ER+ breast cancer, this study examined the pharmacokinetics, clinical uses, mechanisms of action, and problems with treatment resistance. To maximize therapeutic approaches and enhance patient outcomes in the treatment of ER breast cancer, it is imperative to understand these characteristics.
Collapse
Affiliation(s)
- Khushboo Bhutani
- Department of Biotechnology, SRM University, Delhi-NCR, Sonepat, Haryana 131029, India
| | - Suyashi Vishwakarma
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh 201309, India
| | - Priyanka Yadav
- Department of Biotechnology, SRM University, Delhi-NCR, Sonepat, Haryana 131029, India
| | - Manoj Kumar Yadav
- Department of Biotechnology, SRM University, Delhi-NCR, Sonepat, Haryana 131029, India; Department of Biomedical Engineering, SRM University, Sonepat, Haryana 131029, India.
| |
Collapse
|
2
|
Chu Y, Nie Q, Zhou X, Yang J, Fang J, Zhang J. Berberrubine as a novel TrxR inhibitor enhances cisplatin sensitivity in the treatment of non-small cell lung cancer. Bioorg Chem 2025; 158:108329. [PMID: 40056602 DOI: 10.1016/j.bioorg.2025.108329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025]
Abstract
Thioredoxin reductase (TrxR, TXNRD) is an essential enzyme implicated in the processes of cancer development and progression, positioning it as a promising target for cancer therapeutics. In this study, we employed target-based structural screening to identify berberrubine (BRB), a natural product characterized by an unprecedented isoquinoline scaffold that differs from known TrxR inhibitors. Our findings demonstrate that BRB serves as an effective inhibitor of TrxR, both in the context of the purified enzyme and within cancer cells. Since TrxR is highly expressed in non-small cell lung cancer (NSCLC) and is linked to patient prognosis and drug resistance, our results demonstrate, for the first time, that BRB can enhance the sensitivity of cisplatin to impede the proliferation of A549 cells, which was further confirmed in a xenograft model. The primary reason for cisplatin resistance in NSCLC is the DNA repair mechanism of apoptotic tumor cells. Our subsequent mechanistic investigation discovered that BRB selectively inhibits TrxR and impairs the biologically functional thioredoxin, which ultimately inhibits DNA synthesis and repair in cancer cells. Inhibition of TrxR by BRB led to a significant ROS accumulation in A549 cells, which contributed to oxidative stress-mediated apoptosis when used in combination with cisplatin. Our results conclude that BRB is a novel chemical entity of TrxR inhibitor that can increase the effectiveness of cisplatin in slowing down the growth of NSCLC both in vitro and in vivo. This provides a new perspective on the potential application of the combination of the two in the treatment of NSCLC.
Collapse
Affiliation(s)
- Yajun Chu
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Qiuying Nie
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Xiedong Zhou
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Junwei Yang
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Jianguo Fang
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China.; School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing 210094, China..
| | - Junmin Zhang
- School of Pharmacy, and State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, China..
| |
Collapse
|
3
|
Hinterndorfer M, Spiteri VA, Ciulli A, Winter GE. Targeted protein degradation for cancer therapy. Nat Rev Cancer 2025:10.1038/s41568-025-00817-8. [PMID: 40281114 DOI: 10.1038/s41568-025-00817-8] [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] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
Targeted protein degradation (TPD) aims at reprogramming the target specificity of the ubiquitin-proteasome system, the major cellular protein disposal machinery, to induce selective ubiquitination and degradation of therapeutically relevant proteins. Since its conception over 20 years ago, TPD has gained a lot of attention mainly due to improvements in the design of bifunctional proteolysis targeting chimeras (PROTACs) and understanding the mechanisms underlying molecular glue degraders. Today, PROTACs are on the verge of a first clinical approval and recent structural and mechanistic insights combined with technological leaps promise to unlock the rational design of protein degraders, following the lead of lenalidomide and related clinically approved analogues. At the same time, the TPD universe is expanding at a record speed with the discovery of novel modalities beyond molecular glue degraders and PROTACs. Here we review the recent progress in the field, focusing on newly discovered degrader modalities, the current state of clinical degrader candidates for cancer therapy and upcoming design approaches.
Collapse
Affiliation(s)
- Matthias Hinterndorfer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Valentina A Spiteri
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Georg E Winter
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
4
|
Naxerova K. Evolutionary paths towards metastasis. Nat Rev Cancer 2025:10.1038/s41568-025-00814-x. [PMID: 40263543 DOI: 10.1038/s41568-025-00814-x] [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] [Accepted: 03/18/2025] [Indexed: 04/24/2025]
Abstract
The evolution of metastasis in humans is considerably less well understood than the biology of early carcinogenesis. For over a century, clinicians and scientists have been debating whether metastatic potential is the intrinsic property of a cancer, pre-determined by the molecular characteristics of the tumour founder cell, or whether metastatic capacity evolves in a stepwise fashion as the tumour grows, akin to the multistage accumulation of oncogenic alterations that give rise to the first cancer cell. In this Perspective, I examine how genetic analyses of primary tumours and matched metastases can distinguish between these two competing metastasis evolution models, with particular emphasis on the utility of metastatic randomness - a quantitative measure that reflects whether metastases arise from a random selection of primary tumour subclones or whether they are enriched for descendants of privileged lineages that have acquired pro-metastatic traits. Probable metastasis evolution trajectories in tumours with high and low baseline metastatic capacity are discussed, along with the role of seeding rates and selection at different metastatic host sites. Finally, I argue that trailblazing insights into human metastasis biology are immediately possible if we make a concerted effort to apply existing experimental and theoretical tools to the right patient cohorts.
Collapse
Affiliation(s)
- Kamila Naxerova
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Han J, Yuan Y, Zhang J, Hou Y, Xu H, Nie X, Zhao Z, Hou J. Regulatory effect of Wnt signaling on mitochondria in cancer: from mechanism to therapy. Apoptosis 2025:10.1007/s10495-025-02114-z. [PMID: 40257508 DOI: 10.1007/s10495-025-02114-z] [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] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
Cancer is one of the most significant public health challenges in the new millennium, and complex mechanisms are at work to contribute to its pathogenesis and progression. The Wnt signaling pathways, which are crucial conserved cascades involved in embryological development and tissue homeostasis, and mitochondria, the intracellular powerhouses responsible for energy production, calcium and iron homeostasis, as well as mitochondrial apoptosis in eukaryotic cells, have their own mechanisms regulating these pathological processes. In the past decade, accumulating evidence has indicated that Wnt signaling pathways directly regulate mitochondrial biogenesis and function under physiological and pathological conditions. In this review, we systemically summarize the current understanding of how Wnt signaling pathways, particularly the canonical Wnt cascade, regulate mitochondrial fission, respiration, metabolism, and mitochondrial-dependent apoptosis in cancer. In addition, we discuss recent advancements in the research of anticancer agents and related pharmacological mechanisms targeting the signaling transduction of canonical Wnt pathway and/or mitochondrial function. We believe that the combined use of pharmaceuticals targeting Wnt signaling and/or mitochondria with conventional therapies, immunotherapy and targeted therapy based on accurate molecular pathological diagnosis will undoubtedly be the future mainstream direction of personalized cancer treatment, which could benefit more cancer patients.
Collapse
Affiliation(s)
- Jinping Han
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Yimeng Yuan
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Jianhua Zhang
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 475003, Kaifeng, China
| | - Yifan Hou
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Hongtao Xu
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China.
| | - Zhenhua Zhao
- Ma'anshan 86 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 243100, Ma'anshan, China
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 475003, Kaifeng, China
| |
Collapse
|
6
|
Greene G, Zonfa I, Ravasz Regan E. A Boolean network model of hypoxia, mechanosensing and TGF-β signaling captures the role of phenotypic plasticity and mutations in tumor metastasis. PLoS Comput Biol 2025; 21:e1012735. [PMID: 40238833 DOI: 10.1371/journal.pcbi.1012735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
The tumor microenvironment aids cancer progression by promoting several cancer hallmarks, independent of cancer-related mutations. Biophysical properties of this environment, such as the stiffness of the matrix cells adhere to and local cell density, impact proliferation, apoptosis, and the epithelial to mesenchymal transition (EMT). The latter is a rate-limiting step for invasion and metastasis, enhanced in hypoxic tumor environments but hindered by soft matrices and/or high cell densities. As these influences are often studied in isolation, the crosstalk between hypoxia, biomechanical signals, and the classic EMT driver TGF-β is not well mapped, limiting our ability to predict and anticipate cancer cell behaviors in changing tumor environments. To address this, we built a Boolean regulatory network model that integrates hypoxic signaling with a mechanosensitive model of EMT, which includes the EMT-promoting crosstalk of mitogens and biomechanical signals, cell cycle control, and apoptosis. Our model reproduces the requirement of Hif-1α for proliferation, the anti-proliferative effects of strong Hif-1α stabilization during hypoxia, hypoxic protection from anoikis, and hypoxia-driven mechanosensitive EMT. We offer experimentally testable predictions about the effect of VHL loss on cancer hallmarks, with or without secondary oncogene activation. Taken together, our model serves as a predictive framework to synthesize the signaling responses associated with tumor progression and metastasis in healthy vs. mutant cells. Our single-cell model is a key step towards more extensive regulatory network models that cover damage-response and senescence, integrating most cell-autonomous cancer hallmarks into a single model that can, in turn, control the behavior of in silico cells within a tissue model of epithelial homeostasis and carcinoma.
Collapse
Affiliation(s)
- Grant Greene
- Biochemistry and Molecular Biology, College of Wooster, Wooster, Ohio, United States of America
| | - Ian Zonfa
- Biochemistry and Molecular Biology, College of Wooster, Wooster, Ohio, United States of America
| | - Erzsébet Ravasz Regan
- Biochemistry and Molecular Biology, College of Wooster, Wooster, Ohio, United States of America
| |
Collapse
|
7
|
Yin Q, Zhang Y, Xie X, Hou M, Chen X, Ding J. Navigating the future of gastric cancer treatment: a review on the impact of antibody-drug conjugates. Cell Death Discov 2025; 11:144. [PMID: 40188055 PMCID: PMC11972320 DOI: 10.1038/s41420-025-02429-5] [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: 11/05/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Gastric cancer, marked by its high incidence and poor prognosis, demands the urgent development of novel and effective treatment strategies, especially for patients ineligible for surgery or those who have had limited success with chemotherapy, radiotherapy and targeted therapies. Recently, antibody-drug conjugates (ADCs) have become a key area of investigation due to their high specificity and potent antitumor effects. These therapies combine monoclonal antibodies, designed to bind to tumor-specific antigens, with cytotoxic agents that selectively target and destroy malignant cells. ADCs have generated significant interest in clinical trials as a promising approach to improve both treatment efficacy and patient outcomes in gastric cancer. However, their clinical application is not without challenges and limitations that must be addressed. This review discusses the recent progress in the use of ADCs for gastric cancer treatment.
Collapse
Affiliation(s)
- Qingling Yin
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Yanlong Zhang
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Xueqing Xie
- GuiZhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Meijun Hou
- Graduate School, Zunyi Medical University, Zunyi, Guizhou, 563006, China
| | - Xunsheng Chen
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China
| | - Jie Ding
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guiyang, China.
| |
Collapse
|
8
|
Jiang X, Ge X, Huang Y, Xie F, Chen C, Wang Z, Tao W, Zeng S, Lv L, Zhan Y, Bao L. Drug resistance in TKI therapy for hepatocellular carcinoma: Mechanisms and strategies. Cancer Lett 2025; 613:217472. [PMID: 39832650 DOI: 10.1016/j.canlet.2025.217472] [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: 11/17/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Tyrosine kinase inhibitors (TKIs) are such as sorafenib the first-line therapeutic drugs for patients with advanced hepatocellular carcinoma. However, patients with TKI-resistant advanced liver cancer are insensitive to TKI treatment, resulting in limited survival benefits. This paper comprehensively reviewed the mechanisms underlying TKI resistance in hepatocytes, investigating activation of tumor signaling pathways, epigenetic regulation, tumor microenvironment, and metabolic reprogramming. Based on resistance mechanisms, it also reviews preclinical and clinical studies of drug resistance strategies and summarizes targeted therapy combined with immunotherapy currently in investigational clinical trials. Understanding the interactions and clinical studies of these resistance mechanisms offers new hope for improving and prolonging patient survival.
Collapse
Affiliation(s)
- Xue Jiang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Xiaoying Ge
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Yueying Huang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Fangyuan Xie
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Chun Chen
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Zijun Wang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Wanru Tao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Sailiang Zeng
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Lei Lv
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Yangyang Zhan
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Leilei Bao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| |
Collapse
|
9
|
Xue Y, Chen T, Ma Z, Pu X, Xu J, Zhai S, Du X, Ji Y, Simon MC, Zhai W, Xue W. Osalmid sensitizes clear cell renal cell carcinoma to navitoclax through a STAT3/BCL-XL pathway. Cancer Lett 2025; 613:217514. [PMID: 39894195 DOI: 10.1016/j.canlet.2025.217514] [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: 08/22/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common and lethal urinary malignancy characterized by its resistance to apoptosis. Despite the emerging treatment options available for ccRCC, only a small proportion of patients achieve long-term survival benefits. Previous studies have demonstrated that inducing tumor cell senescence, followed by treatment using senolytics, represents a potential strategy for triggering tumor cell apoptosis. However, it remains unclear whether this strategy is suitable for the treatment of ccRCC. Using the whole-genome CRISPR screening database Dependency Map portal (DepMap), we identified ribonucleotide reductase family member 2 (RRM2), which catalyzes the conversion of ribonucleotides to deoxyribonucleotides (dNTPs), as an essential targetable gene for ccRCC. Herein, we report that the combination of the choleretic drug osalmid targeting RRM2 and the senolytic compound navitoclax targeting BCL-XL represents a novel therapeutic approach for ccRCC. Furthermore, we have validated this approach across a panel of human ccRCC cells with different genetic backgrounds and multiple preclinical models, including cell line-derived xenografts (CDX), patient-derived xenografts (PDX), and patient-derived organoids (PDO). Mechanistically, osalmid-mediated inhibition of dNTPs generation induces cellular senescence in ccRCC, concomitant with STAT3 activation and upregulation of BCL-XL, thus rendering these cells vulnerable to navitoclax, which targets the BCL-2 protein family.
Collapse
Affiliation(s)
- Yizheng Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tianyi Chen
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Zehua Ma
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550001, China
| | - Xinyuan Pu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Junyao Xu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Shuanfeng Zhai
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yiyi Ji
- Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, 929 East 57th Street, Chicago, IL, 60637, USA, Howard Hughes Medical Institute
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wei Zhai
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China; Shanghai Immune Therapy Institute State, Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
10
|
Liu M, Guo J, Liu W, Yang Z, Yu F. Dual Targeting of Aurora-A and Bcl-xL Synergistically Reshapes the Immune Microenvironment and Induces Apoptosis in Breast Cancer. Cancer Sci 2025. [PMID: 40159464 DOI: 10.1111/cas.70072] [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: 11/27/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
The Aurora-A kinase inhibitor MLN8237 has shown efficacy in clinical trials for advanced breast cancer; however, its use as a monotherapy is limited by significant side effects and modest efficacy. Therefore, combining MLN8237 with other agents at lower doses may provide a viable alternative. In this study, we evaluated the combination of MLN8237 with the BH3 mimetic ABT263 for the treatment of triple-negative breast cancer (TNBC). We found that this combination significantly suppressed tumor growth and metastasis in immunocompetent syngeneic mouse models, whereas its efficacy was attenuated in immunodeficient xenograft models. Mechanistic studies revealed that the combination enhanced anti-tumor immunity by increasing the presence of CD8+ T cells and NK cells, while reducing the number of immunosuppressive cells in the tumor microenvironment. This shift resulted in elevated levels of IFN-γ and granzyme B, which activated the extrinsic apoptotic pathways in cancer cells. Notably, the combination treatment did not affect tumor cell proliferation but promoted apoptosis with minimal toxicity. Furthermore, the synergistic effect of MLN8237 and ABT263 in inducing intrinsic apoptosis was primarily driven by the inhibition of the AKT-Mcl-1 and Bcl-xL survival pathways in cultured tumor cells. Together, these findings support the MLN8237-ABT263 combination as an effective treatment strategy for TNBC, promoting both immune-mediated extrinsic apoptosis and inactivation of Bcl-xL/Mcl-1-dependent intrinsic anti-apoptotic pathways.
Collapse
Affiliation(s)
- Mingxue Liu
- Department of Ultrasound, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jing Guo
- Department of Ultrasound, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Weiyong Liu
- Department of Ultrasound, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhenye Yang
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fazhi Yu
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
11
|
Endo R, Ueda T, Nagaoki T, Sato Y, Maishi N, Hida K, Harashima H, Nakamura T. Selective vascular disrupting therapy by lipid nanoparticle-mediated Fas ligand silencing and stimulation of STING. Biomaterials 2025; 321:123297. [PMID: 40158445 DOI: 10.1016/j.biomaterials.2025.123297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Although recent therapeutic developments have greatly improved the outcomes of patients with cancer, it remains on ongoing problem, particularly in relation to acquired drug resistance. Vascular disrupting agents (VDAs) directly damage tumor blood vessels, thus promoting drug efficacy and reducing the development of drug resistance; however, their low molecular weight and resulting lack of selectivity for tumor endothelial cells (TECs) lead to side effects that can hinder their practical use. Here, we report a novel tumor vascular disrupting therapy using nucleic acid-loaded lipid nanoparticles (LNPs). We prepared two LNPs: a small interfering RNA (siRNA) against Fas ligand (FasL)-loaded cyclic RGD modified LNP (cRGD-LNP) to knock down FasL in TECs and a stimulator of interferon genes (STING) agonist-loaded LNP to induce systemic type I interferon (IFN) production. The combination therapy disrupted the tumor vasculature and induced broad tumor cell apoptosis within 48 h, leading to rapid and strong therapeutic effects in various tumor models. T cells were not involved in these antitumor effects. Furthermore, the combination therapy demonstrated a significantly superior therapeutic efficacy compared with conventional anti-angiogenic agents and VDAs. RNA sequencing analysis suggested that reduced collagen levels may have been responsible for TEC apoptosis. These findings demonstrated a potential therapeutic method for targeting the tumor vasculature, which may contribute to the development of a new class of anti-cancer drugs.
Collapse
Affiliation(s)
- Rikito Endo
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Tomoki Ueda
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Takumi Nagaoki
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Nako Maishi
- Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, 060-8586, Japan
| | - Kyoko Hida
- Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, 060-8586, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan.
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan; Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan.
| |
Collapse
|
12
|
Giri S, Lamichhane G, Pandey J, Khadayat R, K. C. S, Devkota HP, Khadka D. Immune Modulation and Immunotherapy in Solid Tumors: Mechanisms of Resistance and Potential Therapeutic Strategies. Int J Mol Sci 2025; 26:2923. [PMID: 40243502 PMCID: PMC11989189 DOI: 10.3390/ijms26072923] [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: 02/04/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Understanding the modulation of specific immune cells within the tumor microenvironment (TME) offers new hope in cancer treatments, especially in cancer immunotherapies. In recent years, immune modulation and resistance to immunotherapy have become critical challenges in cancer treatments. However, novel strategies for immune modulation have emerged as promising approaches for oncology due to the vital roles of the immunomodulators in regulating tumor progression and metastasis and modulating immunological responses to standard of care in cancer treatments. With the progress in immuno-oncology, a growing number of novel immunomodulators and mechanisms are being uncovered, offering the potential for enhanced clinical immunotherapy in the near future. Thus, gaining a comprehensive understanding of the broader context is essential. Herein, we particularly summarize the paradoxical role of tumor-related immune cells, focusing on how targeted immune cells and their actions are modulated by immunotherapies to overcome immunotherapeutic resistance in tumor cells. We also highlight the molecular mechanisms employed by tumors to evade the long-term effects of immunotherapeutic agents, rendering them ineffective.
Collapse
Affiliation(s)
- Suman Giri
- Asian College for Advance Studies, Purbanchal University, Satdobato, Lalitpur 44700, Nepal;
| | - Gopal Lamichhane
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Jitendra Pandey
- Department of Chemistry, University of Hawai’i at Manoa, 2545 McCarthy Mall, Honolulu, HI 96822, USA;
| | - Ramesh Khadayat
- Patan Hospital, Patan Academic of Health Sciences, Lagankhel, Lalitpur 44700, Nepal;
| | - Sindhu K. C.
- Department of Pharmacology, Chitwan Medical College, Tribhuwan University, Bharatpur-05, Chitwan 44200, Nepal;
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Oehonmachi 5-1, Chuo-ku, Kumamoto 862-0973, Japan;
- Headquarters for Admissions and Education, Kumamoto University, Kurokami, 2-39-1, Chuo-ku, Kumamoto 860-8555, Japan
| | - Dipendra Khadka
- NADIANBIO Co., Ltd., Wonkwang University School of Medicine, Business Incubation Center R201-1, Iksan 54538, Jeonbuk, Republic of Korea
- KHAS Health Pvt. Ltd., Dhangadhi-04, Kailali 10910, Nepal
| |
Collapse
|
13
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [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: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
14
|
Huang L, Zeng X, Xiao K, Tang S, Sun K. Silencing Livin gene expression by RNA interference enhanced the chemotherapeutic sensitivity of drug-resistant osteosarcoma cells to doxorubicin. Acta Histochem 2025; 127:152249. [PMID: 40121921 DOI: 10.1016/j.acthis.2025.152249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 03/08/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Osteosarcoma is one of the most common malignant tumors in children and adolescents. It occurs in the metaphysis of long bones and is a type of aggressive malignant tumor. Although there are treatment methods such as surgery and chemotherapy, the mortality and disability of osteosarcoma patients are still high. With the emergence of more and more chemotherapy resistance, it is necessary to find new therapies to improve the chemotherapy sensitivity of osteosarcoma. METHODS Drug-resistant MG-63 and U2OS cell strain was established in vitro by continuous exposure of human osteosarcoma cells to doxorubicin at gradually increasing concentrations,then determined for resistance index to doxorubicin by MTT method,for transcriptions of Livin mRNA by real-time polymerase chain reaction(RT⁃PCR),and for expressions of Livin proteins by Western blot.The technology of gene recombination was used to construct the eukaryotic expression vector pSilencer3.1-H1 neo-Livin. Then the pSilencer3.1-H1 neo-Livin was transfected into drug-resistant MG-63 cell by using Lipofectmine 2000. Expressions of Livin mRNA and protein in the transfected cells were respectively measured by RT-PCR and Western blot. The distribution of cell cycle phase and apoptosis were determined by flow cytometry. The analysis of chemotherapeutic sensitivity of drug-resistant MG-63 cell to doxorubicin was performed by MTT. RESULTS The recombinant eukaryotic expression vector pSilencer3.1-H1 neo-Livin was successfully constructed. The result of inverted microscope revealed that the drug-resistant MG-63 cell were irregularity and morphological diversity. Compared with those in osteosarcoma cells,the transcription levels of Livin mRNA and protein in drug-resistant osteosarcoma cell increased(P<0.05).The flow cytometry analysis showed there was higher percentage of apoptosis in transfected drug-resistant MG-63 cell. Compared with control groups,the expression of Livin mRNA and protein were both significantly decreased in the transfected drug-resistant osteosarcomacell(P<0.05). We also observed that suppression of Livin expression in osteosarcoma cells increased their chemosensitivity to doxorubicin. CONCLUSION This study showed that Livin shRNA inhibited the proliferation level and increased the sensitivity of drug-resistant osteosarcoma cell to doxorubicin, suggested that Livin is involved in drug resistance of human osteosarcoma and may serve as a promising therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Lei Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Xiaobin Zeng
- Department of Basic nursing, Nanchang Health School, Nanchang 330006, China; Department of Basic nursing, Nanchang Health Vocational And Technical College, Nanchang 330006, China
| | - Kaimin Xiao
- Department of Orthopedic Surgery, the People's Hospital of Jishui County, Jian 344500, China
| | - Sen Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Kuo Sun
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
15
|
He D, Yang Z, Zhang T, Luo Y, Peng L, Yan J, Qiu T, Zhang J, Qin L, Liu Z, Sun M. Multi-omics and machine learning-driven CD8 + T cell heterogeneity score for head and neck squamous cell carcinoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102413. [PMID: 40027882 PMCID: PMC11869859 DOI: 10.1016/j.omtn.2024.102413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/03/2024] [Indexed: 03/05/2025]
Abstract
The heterogeneity of head and neck squamous cell carcinoma (HNSCC) poses a significant challenge to treatment, underscoring the urgent need for more precise and personalized therapeutic approaches. CD8+ T cells, integral components of the tumor immune microenvironment, have emerged as key targets for immunotherapy. Our research has established a correlation between a decrease in CD8+ T cell score and a poor clinical prognosis, highlighting the prognostic value of this biomarker. By analyzing the gene expression related to CD8+ T cells, we have differentiated HNSCC into cold and hot tumor subtypes, uncovering disparities in clinical prognosis and responses to immunotherapy. Utilizing eight machine learning methods, we identified the key gene OLR1. Single-cell analysis of HNSCC tissues and peripheral blood, along with spatial transcriptome analysis, revealed that OLR1 predominantly functions in macrophages, modulating the immune microenvironment of HNSCC. The expression level of OLR1 may serve as a predictive marker for immunotherapy responses. Moreover, drug sensitivity analysis and molecular docking studies have indicated that simvastatin and pazopanib are potential inhibitors of OLR1. These findings suggest that simvastatin and pazopanib could open up innovative potential therapeutic avenues for individuals with HNSCC.
Collapse
Affiliation(s)
- Di He
- Department of Oral and Maxillofacial Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhan Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Tian Zhang
- Department of Oral and Maxillofacial Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yaxian Luo
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Lianjie Peng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jiatao Yan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Tao Qiu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jingyu Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Luying Qin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Zhichao Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Mouyuan Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang Province, China
| |
Collapse
|
16
|
Zhou M, Han R, Xu W, Hao X, Peng Y, Tang Y, Sun P, Tang T, Wu J, Xiang D. Biomimetic Atorvastatin Self-Assembled Nanomedicine Inhibits the Cyclooxygenase-2/Prostaglandin E2 Pathway Enhanced Photothermal and Antitumor Immunity. Biomater Res 2025; 29:0149. [PMID: 40040956 PMCID: PMC11876541 DOI: 10.34133/bmr.0149] [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: 12/23/2024] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Cancer continues to pose remarkable medical challenges worldwide. While current cancer therapies can lead to initial clinical improvement, they are often followed by recurrence, metastasis, and drug resistance, underscoring the urgent need for innovative treatment strategies. Atorvastatin calcium (AC), a widely used lipid-lowering and anti-inflammation drug in the clinic, has shown antitumor potential. To further improve the antitumor efficacy, we developed self-assembled AC and polydopamine (PDA) nanoparticles whose surface was coated with macrophage membranes (CM) as a biomimetic drug delivery system [AC@PDA@CM (APM)]. APM showed high drug-loading capacity, excellent stability, excellent bioavailability, and tumor-targeting ability, ultimately achieving photothermal synergistic cancer immunotherapy. Our findings indicate that APM efficiently delivers AC to tumor sites while leveraging photothermal therapy (PTT) to enhance local tumor ablation and antitumor immune effect. Notably, APM mitigates tumor immunosuppression triggered by PTT through AC, suppressing the COX-2/PGE2 pathway and immune evasion signal CD47. Furthermore, APM notably reduced nonspecific distribution and side effects, which is conducive to ensuring the safety level of medication. This integrated approach boosts therapeutic efficacy and highlights the potential of APM as a multifunctional agent for cancer therapy, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Min Zhou
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Ruyue Han
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Xinyan Hao
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Yanjin Peng
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Yucheng Tang
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Pengcheng Sun
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital,
Central South University, Changsha 410011, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, China
- Institute of Clinical Pharmacy, Central South University, Changsha 410011, China
| |
Collapse
|
17
|
Ligero M, El Nahhas OSM, Aldea M, Kather JN. Artificial intelligence-based biomarkers for treatment decisions in oncology. Trends Cancer 2025; 11:232-244. [PMID: 39814650 DOI: 10.1016/j.trecan.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
The development of new therapeutic strategies such as immune checkpoint inhibitors (ICIs) and targeted therapies has increased the complexity of the treatment landscape for solid tumors. At the current rate of annual FDA approvals, the potential treatment options could increase by tenfold over the next 5 years. The cost of personalized medicine technologies limits its accessibility, thus increasing socioeconomic disparities in the treated population. In this review we describe artificial intelligence (AI)-based solutions - including deep learning (DL) methods for routine medical imaging and large language models (LLMs) for electronic health records (EHRs) - to support cancer treatment decisions with cost-effective biomarkers. We address the current limitations of these technologies and propose the next steps towards their adoption in routine clinical practice.
Collapse
Affiliation(s)
- Marta Ligero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany
| | - Mihaela Aldea
- Department of Cancer Medicine, Institut Gustave Roussy, Université Paris-Saclay, F-94805, Villejuif, France; Thoracic Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Dresden University of Technology (TUD), Dresden, Germany; Department of Medicine I, University Hospital Dresden, Dresden, Germany; Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
18
|
Guo Y, Li Y, Li J, Cai H, Liu K, Duan D, Zhang W, Han G, Zhao Y. Controlled Inflammation Drives Neutrophil-Mediated Precision Drug Delivery in Heterogeneous Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411307. [PMID: 39799561 PMCID: PMC11923894 DOI: 10.1002/advs.202411307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/05/2024] [Indexed: 01/15/2025]
Abstract
Tumor heterogeneity remains a formidable obstacle in targeted cancer therapy, often leading to suboptimal treatment outcomes. This study presents an innovative approach that harnesses controlled inflammation to guide neutrophil-mediated drug delivery, effectively overcoming the limitations imposed by tumor heterogeneity. By inducing localized inflammation within tumors using lipopolysaccharide, it significantly amplify the recruitment of drug-laden neutrophils to tumor sites, irrespective of specific tumor markers. This strategy not only enhances targeted drug delivery but also triggers the release of neutrophil extracellular traps, further potentiating the anti-tumor effect. Crucially, this study demonstrates that potential systemic inflammatory responses can be effectively mitigated through neutrophil transfusion, ensuring the safety and clinical viability of this approach. In a murine breast cancer model, the method significantly impedes tumor growth compared to conventional treatments. This work offers a versatile strategy for precise drug delivery across diverse tumor types. The findings pave the way for more effective and broadly applicable cancer treatments, potentially addressing the long-standing challenge of tumor heterogeneity.
Collapse
Affiliation(s)
- Yunfei Guo
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, P. R. China
| | - Yiming Li
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, P. R. China
| | - Jianmin Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, P. R. China
| | - Haoran Cai
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, P. R. China
| | - Kangkang Liu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, P. R. China
| | - Dengyi Duan
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, P. R. China
| | - Wenyi Zhang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, P. R. China
| | - Gang Han
- Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Yang Zhao
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, P. R. China
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, P. R. China
- Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| |
Collapse
|
19
|
Zheng H, Zhang K, Piao J, Mu C, Xie X, Cheng M, Yue T, Sun J, Li B, Wei Y, Zheng H, Jiang L, Habiballah DN, Li F. In situ valence-transited arsenic nanosheets for multi-modal therapy of colorectal cancer. Nat Commun 2025; 16:2088. [PMID: 40025024 PMCID: PMC11873056 DOI: 10.1038/s41467-025-57376-7] [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: 09/04/2023] [Accepted: 02/12/2025] [Indexed: 03/04/2025] Open
Abstract
Late-stage and advanced colorectal cancer (CRC) often prove to be resistant to current treatment regimens, due to the evolving tumor microenvironment. Chemotherapy-dominated multi-modal therapeutic strategies based on the specific CRC microenvironment open a new horizon for eradicating colorectal tumors. Here, in situ valence-transited arsenic nanosheets are developed as a multi-modal therapeutic platform by responding to the H2S-enriched CRC microenvironment. Carrier-free pegylated nanosheets of pentavalent arsenic (AsV), aminooxyacetic acid (AOAA), and copper ion (Cu2+) are innovatively self-assembled via coordination with high loading content and good stability. AsV in pegylated arsenic nanosheets (CAA-PEG NSs) is rapidly released and reduced to trivalent arsenic (AsIII) to exert its chemotherapy in the local tumor. Furthermore, the immunosuppressive microenvironment is thoroughly remodeled via H2S depletion of AsV to AsIII conversion and impairment of H2S production by AOAA. Additionally, the in situ produced ultrasmall CuS nanoparticles exhibit photothermal activity against CRC under the guidance of photoacoustic imaging. This multi-modal therapeutic strategy, dominated by chemotherapy, completely inhibits CRC progression and prevents its relapse.
Collapse
Affiliation(s)
- Hongyue Zheng
- Libraries of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ke Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jigang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chaofeng Mu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaowei Xie
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengying Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tianxiang Yue
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiang Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yinghui Wei
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hangsheng Zheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lai Jiang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | | | - Fanzhu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
20
|
Vo DK, Trinh KTL. Polymerase Chain Reaction Chips for Biomarker Discovery and Validation in Drug Development. MICROMACHINES 2025; 16:243. [PMID: 40141854 PMCID: PMC11944077 DOI: 10.3390/mi16030243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025]
Abstract
Polymerase chain reaction (PCR) chips are advanced, microfluidic platforms that have revolutionized biomarker discovery and validation because of their high sensitivity, specificity, and throughput levels. These chips miniaturize traditional PCR processes for the speed and precision of nucleic acid biomarker detection relevant to advancing drug development. Biomarkers, which are useful in helping to explain disease mechanisms, patient stratification, and therapeutic monitoring, are hard to identify and validate due to the complexity of biological systems and the limitations of traditional techniques. The challenges to which PCR chips respond include high-throughput capabilities coupled with real-time quantitative analysis, enabling researchers to identify novel biomarkers with greater accuracy and reproducibility. More recent design improvements of PCR chips have further expanded their functionality to also include digital and multiplex PCR technologies. Digital PCR chips are ideal for quantifying rare biomarkers, which is essential in oncology and infectious disease research. In contrast, multiplex PCR chips enable simultaneous analysis of multiple targets, therefore simplifying biomarker validation. Furthermore, single-cell PCR chips have made it possible to detect biomarkers at unprecedented resolution, hence revealing heterogeneity within cell populations. PCR chips are transforming drug development, enabling target identification, patient stratification, and therapeutic efficacy assessment. They play a major role in the development of companion diagnostics and, therefore, pave the way for personalized medicine, ensuring that the right patient receives the right treatment. While this tremendously promising technology has exhibited many challenges regarding its scalability, integration with other omics technologies, and conformity with regulatory requirements, many still prevail. Future breakthroughs in chip manufacturing, the integration of artificial intelligence, and multi-omics applications will further expand PCR chip capabilities. PCR chips will not only be important for the acceleration of drug discovery and development but also in raising the bar in improving patient outcomes and, hence, global health care as these technologies continue to mature.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea;
| | - Kieu The Loan Trinh
- Bionano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
21
|
Momoli C, Costa B, Lenti L, Tubertini M, Parenti MD, Martella E, Varchi G, Ferroni C. The Evolution of Anticancer 3D In Vitro Models: The Potential Role of Machine Learning and AI in the Next Generation of Animal-Free Experiments. Cancers (Basel) 2025; 17:700. [PMID: 40002293 PMCID: PMC11853635 DOI: 10.3390/cancers17040700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The development of anticancer therapies has increasingly relied on advanced 3D in vitro models, which more accurately mimic the tumor microenvironment compared to traditional 2D cultures. This review describes the evolution of these 3D models, highlighting significant advancements and their impact on cancer research. We discuss the integration of machine learning (ML) and artificial intelligence (AI) in enhancing the predictive power and efficiency of these models, potentially reducing the dependence on animal testing. ML and AI offer innovative approaches for analyzing complex data, optimizing experimental conditions, and predicting therapeutic outcomes with higher accuracy. By leveraging these technologies, the next generation of 3D in vitro models could revolutionize anticancer drug development, offering effective alternatives to animal experiments.
Collapse
Affiliation(s)
| | | | | | | | | | - Elisa Martella
- Institute for the Organic Synthesis and Photoreactivity—Italian National Research Council, 40129 Bologna, Italy; (C.M.); (B.C.); (L.L.); (M.T.); (M.D.P.); (C.F.)
| | - Greta Varchi
- Institute for the Organic Synthesis and Photoreactivity—Italian National Research Council, 40129 Bologna, Italy; (C.M.); (B.C.); (L.L.); (M.T.); (M.D.P.); (C.F.)
| | | |
Collapse
|
22
|
Pan Z, Qian Y, Wang Y, Zhang T, Song X, Ding H, Li R, Zhang Y, Wang Z, Wang H, Xia W, Wei L, Xu L, Dong G, Jiang F. STAT3 Inhibition Prevents Adaptive Resistance and Augments NK Cell Cytotoxicity to KRAS G12C Inhibitors in Nonsmall Cell Lung Cancer. Cancer Sci 2025. [PMID: 39939568 DOI: 10.1111/cas.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/14/2025] Open
Abstract
KRASG12C inhibitors exhibit conspicuous clinical response in KRASG12C-mutant lung cancer, yet adaptive resistance, the rapid onset of intrinsic resistance, dampens their therapeutic success. Rational combination strategies could tackle this challenging problem. A high-throughput screening of a pharmacological library with 423 compounds revealed that napabucasin, a signal transducer and activator of transcription 3 (STAT3) inhibitor, synergistically potentiated the growth inhibition effect of the KRASG12C inhibitor sotorasib in sensitive and resistant KRASG12C NSCLC cell lines. Functional assays further revealed that the coordinated targeting of KRAS with STAT3 improved the inhibitory effect on tumor growth and augmented the infiltration and activation of natural killer (NK) cells within the tumor microenvironment. Mechanistically, KRASG12C inhibition induced compensatory activation of STAT3, contingent on concomitant suppression of downstream ERK signaling, abrogated by napabucasin. Moreover, we unveiled and verified the binding site of phosphorylated STAT3 at the HLA-B promoter, an inhibitor ligand for NK cells. Our study dissected an unknown mechanism of adaptive resistance to KRASG12C inhibitors, with the STAT3 activation sustaining the regrowth of tumor cells under KRAS inhibition and up-regulating HLA-B transcription to dampen the cytotoxicity of infiltrated NK cells.
Collapse
Affiliation(s)
- Zehao Pan
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
- Department of Pathology, Jiangsu Cancer Hospital, Nanjing, P. R. China
| | - Yuxian Qian
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Yajing Wang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
- Department of Pathology, Jiangsu Cancer Hospital, Nanjing, P. R. China
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, P. R. China
| | - Te Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, P. R. China
- Tianjin Institute of Coloproctology, Tianjin, P. R. China
| | - Xuming Song
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Hanling Ding
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Rutao Li
- Department of Thoracic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, P. R. China
| | - Yijian Zhang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Zi Wang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Hui Wang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Wenjie Xia
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Lei Wei
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
- Department of Pathology, Jiangsu Cancer Hospital, Nanjing, P. R. China
| | - Lin Xu
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, P. R. China
| | - Gaochao Dong
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| | - Feng Jiang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, P. R. China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, P. R. China
| |
Collapse
|
23
|
Arandhara A, Bhuyan P, Das BK. Exploring lung cancer microenvironment: pathways and nanoparticle-based therapies. Discov Oncol 2025; 16:159. [PMID: 39934547 DOI: 10.1007/s12672-025-01902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Lung cancer stands out as a significant global health burden, with staggering incidence and mortality rates primarily linked to smoking and environmental carcinogens. The tumor microenvironment (TME) emerges as a critical determinant of cancer progression and treatment outcomes, comprising a complex interplay of cells, signaling molecules, and extracellular matrix. Through a comprehensive literature review, we elucidate current research trends and therapeutic prospects, aiming to advance our understanding of TME modulation strategies and their clinical implications for lung cancer treatment. Dysregulated immune responses within the TME can facilitate tumor evasion, limiting the efficacy of immune checkpoint inhibitors (ICI). Consequently, TME modulation strategies have become potential avenues to enhance therapeutic responses. However, conventional TME-targeted therapies often face challenges. In contrast, nanoparticle (NP)-based therapies offer promising prospects for improved drug delivery and reduced toxicity, leveraging the enhanced permeability and retention (EPR) effect. Despite NP design and delivery advancements, obstacles like poor tumor cell uptake and off-target effects persist, necessitating further optimization. This review underscores the pivotal role of TME in lung cancer management, emphasizing the synergistic potential of immunotherapy and nano-therapy.
Collapse
Affiliation(s)
- Arunabh Arandhara
- Assam Pharmacy Institute, Titabar, Amgurikhat, Jorhat, Assam, 785632, India
| | - Pallabi Bhuyan
- School of Pharmacy, The Assam Kaziranga University, Koraikhowa, Jorhat, Assam, 785006, India
| | - Bhrigu Kumar Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam, 781017, India.
| |
Collapse
|
24
|
Santoni M, Mollica V, Rizzo A, Massari F. Dynamics of resistance to immunotherapy and TKI in patients with advanced renal cell carcinoma. Cancer Treat Rev 2025; 133:102881. [PMID: 39799795 DOI: 10.1016/j.ctrv.2025.102881] [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: 10/13/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
Immune-based combinations are the cornerstone of the first-line treatment of metastatic renal cell carcinoma patients, leading to outstanding outcomes. Nevertheless, primary resistance and disease progression is a critical clinical challenge. To properly address this issue, it is pivotal to understand the mechanisms of resistance to immunotherapy and tyrosine kinase inhibitors, that tumor eventually develop under treatment. In this review of the literature, we aim at exploring resistance mechanisms arising in patients treated with first-line immune-based combinations in order to understand the biological pattern that should be investigated to overcome them. In more detail, mechanisms of resistance to nivolumab and pembrolizumab are divided into intrinsic to cancer cells and extrinsic (stromal or immune cells). Regarding axitinib, the increased expression of Nuclear protein 1 (NUPR1) or decreased levels of insulin receptor (INSR) characterize resistant cells. The secretion of non-VEGF pro-angiogenic factors, such as PDGF-BB, IL-1β, MMP-9, Gro-α, IL-8, IL-6, and CCL-2, can lead to resistance to cabozantinib. The reactivation of pathways previously targeted by lenvatinib or the activation of alternative pathways, such as EGFR-PAK2-ERK pathway, underlie the development of resistance to lenvatinib. Exploring resistance mechanism that arise during first-line therapy can lead to the development of treatment strategy able to overcome them in order to improve duration of response and patients outcomes.
Collapse
Affiliation(s)
- Matteo Santoni
- Medical Oncology Unit, Macerata Hospital, Macerata, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy.
| |
Collapse
|
25
|
Nabipour H, Aliakbari F, Volkening K, Strong MJ, Rohani S. Development of metal-organic framework biocomposites from chitosan as drug delivery vehicles: In vitro evaluation on HeLa and SH-SY5Y cell lines. Int J Biol Macromol 2025; 291:138878. [PMID: 39701252 DOI: 10.1016/j.ijbiomac.2024.138878] [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: 06/17/2024] [Revised: 11/22/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
In modern times, achieving precise drug delivery through a safe and stable carrier remains a significant challenge. In this study, we synthesized a novel ligand based on a guanazole Schiff base and subsequently developed new metal-organic framework (MOF) named UWO-1 through a reaction involving zinc acetate. At the same time, curcumin (CUR) was loaded onto the newly synthesized UWO-1. The formation of UWO-1 and CUR loading were confirmed through various spectral analyses, including FT-IR, SEM, PXRD, XPS, TGA, zeta potential, DLS, and BET. The specific BET surface area of the UWO-1 is approximately 1456.50 m2/g. The resulting drug delivery system exhibited a drug loading of 30.7 % and an encapsulation efficiency of 94.9 %. CUR@UWO-1 demonstrated rapid drug release, leading to the application of a chitosan (CS) coating to achieve controlled and delayed intravenous delivery, resulting in the creation of CS/CUR@UWO-1 biocomposite microspheres. The cumulative release profile of CUR from CS/CUR@UWO-1 was evaluated in phosphate-buffered saline (PBS) at pH 5.0 and 7.4 over 18 h, revealing release percentages of 66.99 % and 47.18 %, respectively. The release kinetics were studied and found to closely follow the Higuchi model, which was determined to be the best fit. Furthermore, the material's cytotoxicity and anticancer activity were assessed by measuring mitochondrial metabolic activity in vitro. Additionally, this nanosystem was able to internalize the drug into the cells. The combined advantages of enhanced drug loading and sustained pH-responsive release highlight the potential of the MOF-based biocomposite as a versatile drug delivery system for therapeutic applications.
Collapse
Affiliation(s)
- Hafezeh Nabipour
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada
| | - Farhang Aliakbari
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Department of Bioprocess Engineering, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Kathryn Volkening
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Michael J Strong
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Sohrab Rohani
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada.
| |
Collapse
|
26
|
Belmontes B, Slemmons KK, Su C, Liu S, Policheni AN, Moriguchi J, Tan H, Xie F, Aiello DA, Yang Y, Lazaro R, Aeffner F, Rees MG, Ronan MM, Roth JA, Vestergaard M, Cowland S, Andersson J, Sarvary I, Chen Q, Sharma P, Lopez P, Tamayo N, Pettus LH, Ghimire-Rijal S, Mukund S, Allen JR, DeVoss J, Coxon A, Rodon J, Ghiringhelli F, Penel N, Prenen H, Glad S, Chuang CH, Keyvanjah K, Townsley DM, Butler JR, Bourbeau MP, Caenepeel S, Hughes PE. AMG 193, a Clinical Stage MTA-Cooperative PRMT5 Inhibitor, Drives Antitumor Activity Preclinically and in Patients with MTAP-Deleted Cancers. Cancer Discov 2025; 15:139-161. [PMID: 39282709 PMCID: PMC11726016 DOI: 10.1158/2159-8290.cd-24-0887] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/20/2024] [Accepted: 09/10/2024] [Indexed: 01/30/2025]
Abstract
One of the most robust synthetic lethal interactions observed in multiple functional genomic screens has been the dependency on protein arginine methyltransferase 5 (PRMT5) in cancer cells with MTAP deletion. We report the discovery of the clinical stage MTA-cooperative PRMT5 inhibitor AMG 193, which preferentially binds PRMT5 in the presence of MTA and has potent biochemical and cellular activity in MTAP-deleted cells across multiple cancer lineages. In vitro, PRMT5 inhibition induces DNA damage, cell cycle arrest, and aberrant alternative mRNA splicing in MTAP-deleted cells. In human cell line and patient-derived xenograft models, AMG 193 induces robust antitumor activity and is well tolerated with no impact on normal hematopoietic cell lineages. AMG 193 synergizes with chemotherapies or the KRAS G12C inhibitor sotorasib in vitro and combination treatment in vivo substantially inhibits tumor growth. AMG 193 is demonstrating promising clinical activity, including confirmed partial responses in patients with MTAP-deleted solid tumors from an ongoing phase 1/2 study. Significance: AMG 193 preferentially inhibits the growth of MTAP-deleted tumor cells by inhibiting PRMT5 when in complex with MTA, thus sparing MTAP wild-type normal cells. AMG 193 shows promise as a targeted therapy in a clinically defined patient population.
Collapse
Affiliation(s)
| | | | - Chun Su
- Amgen Research, South San Francisco, California
| | - Siyuan Liu
- Amgen Research, Thousand Oaks, California
| | | | | | - Hong Tan
- Amgen Research, Thousand Oaks, California
| | - Fang Xie
- Amgen Research, South San Francisco, California
| | | | | | | | | | - Matthew G. Rees
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | | | | | | | | | | | - Qing Chen
- Amgen Research, Thousand Oaks, California
| | | | | | | | | | | | | | | | | | | | | | | | | | - Hans Prenen
- Universitair Ziekenhuis Antwerpen, Edegem, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Peng H, Chao Z, Wang Z, Hao X, Xi Z, Ma S, Guo X, Zhang J, Zhou Q, Qu G, Gao Y, Luo J, Wang Z, Wang J, Li L. Biomechanics in the tumor microenvironment: from biological functions to potential clinical applications. Exp Hematol Oncol 2025; 14:4. [PMID: 39799341 PMCID: PMC11724500 DOI: 10.1186/s40164-024-00591-7] [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: 09/16/2024] [Accepted: 12/10/2024] [Indexed: 01/15/2025] Open
Abstract
Immune checkpoint therapies have spearheaded drug innovation over the last decade, propelling cancer treatments toward a new era of precision therapies. Nonetheless, the challenges of low response rates and prevalent drug resistance underscore the imperative for a deeper understanding of the tumor microenvironment (TME) and the pursuit of novel targets. Recent findings have revealed the profound impacts of biomechanical forces within the tumor microenvironment on immune surveillance and tumor progression in both murine models and clinical settings. Furthermore, the pharmacological or genetic manipulation of mechanical checkpoints, such as PIEZO1, DDR1, YAP/TAZ, and TRPV4, has shown remarkable potential in immune activation and eradication of tumors. In this review, we delved into the underlying biomechanical mechanisms and the resulting intricate biological meaning in the TME, focusing mainly on the extracellular matrix, the stiffness of cancer cells, and immune synapses. We also summarized the methodologies employed for biomechanical research and the potential clinical translation derived from current evidence. This comprehensive review of biomechanics will enhance the understanding of the functional role of biomechanical forces and provide basic knowledge for the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Hao Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zheng Chao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zefeng Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaodong Hao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zirui Xi
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Sheng Ma
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Xiangdong Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Junbiao Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Qiang Zhou
- Department of Urology, Qinghai University Affiliated Hospital, Qinghai University Medical College, Xining, 810001, Qinghai, China
| | - Guanyu Qu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Yuan Gao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
- Taikang Tongji (Wuhan) Hospital, 420060, Wuhan, China.
| | - Jing Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
| | - Le Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
| |
Collapse
|
28
|
Yang Y, Yu S, Liu W, Zhuo Y, Qu C, Zeng Y. Ferroptosis-related signaling pathways in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:1. [PMID: 39935430 PMCID: PMC11813627 DOI: 10.20517/cdr.2024.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
Ferroptosis is an iron-dependent form of programmed cell death induced by lipid peroxidation. This process is regulated by signaling pathways associated with redox balance, iron metabolism, and lipid metabolism. Cancer cells' increased iron demand makes them especially susceptible to ferroptosis, significantly influencing cancer development, therapeutic response, and metastasis. Recent findings indicate that cancer cells can evade ferroptosis by downregulating key signaling pathways related to this process, contributing to drug resistance. This underscores the possibility of modulating ferroptosis as an approach to counteract drug resistance and enhance therapeutic efficacy. This review outlines the signaling pathways involved in ferroptosis and their interactions with cancer-related signaling pathways. We also highlight the current understanding of ferroptosis in cancer drug resistance, offering insights into how targeting ferroptosis can provide novel therapeutic approaches for drug-resistant cancers. Finally, we explore the potential of ferroptosis-inducing compounds and examine the challenges and opportunities for drug development in this evolving field.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Simin Yu
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wanyao Liu
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Yi Zhuo
- First Clinical Department of Changsha Medical University, Changsha 410219, Hunan, China
| | - Chunrun Qu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
29
|
Mattoo S, Arora M, Sharma P, Pore SK. Targeting mammalian N-end rule pathway for cancer therapy. Biochem Pharmacol 2025; 231:116684. [PMID: 39613115 DOI: 10.1016/j.bcp.2024.116684] [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: 08/29/2024] [Revised: 11/12/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Regulated protein degradation plays a crucial role in maintaining proteostasis along with protein refolding and compartmentalisation which collectively control biological functions. The N-end rule pathway is a major ubiquitin-dependent protein degradation system. The short-lived protein substrates containing destabilizing amino acid residues (N-degrons) are recognized by E3 ubiquitin ligases containing UBR box domains (N-recognin) for degradation. The dysregulated pathway fails to maintain the metabolic stability of the substrate proteins which leads to diseases. The mammalian substrates of this pathway are involved in many hallmarks of cancer such as resisting cell death, evading growth suppression, chromosomal instability, angiogenesis, and deregulation of cellular metabolism. Besides, mutations in E3 N-recognin have been detected in human cancers. In this review, we discuss the mammalian N-end rule pathway components, functions, and mechanism of degradation of substrates, and their implications in cancer pathogenesis. We also discuss the impact of pharmacological and genetic inhibition of this pathway component on cancer cells and chemoresistance. We further highlight how this pathway can be manipulated for selective protein degradation; for instance, using PROTAC technique. The challenges and future perspectives to utilize this pathway as a drug target for cancer therapy are also discussed.
Collapse
Affiliation(s)
- Shria Mattoo
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201311, India
| | - Muskaan Arora
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201311, India
| | - Priyanka Sharma
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Noida 201311, India
| | - Subrata Kumar Pore
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Uttar Pradesh, Noida 201311, India.
| |
Collapse
|
30
|
Pallathadka H, Jabir M, Rasool KH, Hanumanthaiah M, Sharma N, Pramanik A, Rab SO, Jawad SF, Oghenemaro EF, Mustafa YF. siRNA-based therapy for overcoming drug resistance in human solid tumours; molecular and immunological approaches. Hum Immunol 2025; 86:111221. [PMID: 39700968 DOI: 10.1016/j.humimm.2024.111221] [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: 08/10/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
RNA interference (RNAi) is a primordial biological process that protects against external intrusion. SiRNA has the potential to selectively silence disease-related genes in a sequence-specific way, thus offering a promising therapeutic approach. The efficacy of siRNA-based therapies in cancer treatment has gained significant recognition due to multiple studies demonstrating its ability to effectively suppress cancer cells' growth and multiplication. Moreover, siRNA-based medicines have shown considerable promise in enhancing the sensitivity of cancer cells to chemotherapy and other treatment methods by suppressing genes that play a role in the development of drug resistance. Exploring and identifying functional genes linked to cancer cell characteristics and drug resistance is crucial for developing effective siRNAs for cancer treatment and advancing targeted and personalized therapeutics. Targeting and silencing genes in charge of resistance mechanisms, such as those involved in drug efflux, cell survival, or DNA repair, is possible with siRNA therapy in the context of drug resistance, especially cancer. Through inhibiting these genes, siRNA therapy can prevent resistance and restore the efficacy of traditional medications. This review addresses the potential of siRNAs in addressing drug resistance in human tumours, opening up new possibilities in cancer therapy. This review article offers a non-systematic summary of how different siRNA types contribute to cancer cells' treatment resistance. Using pertinent keywords, sources were chosen from reliable databases, including PubMed, Scopus, and Google Scholar. The review covered essential papers in this area and those that mainly addressed the function of siRNA in drug resistance. The articles examined in connection with the title of this review were primarily published from 2020 onward and are based on in vitro studies. Furthermore, this article examines the potential barriers and prospective perspectives of siRNA therapies.
Collapse
Affiliation(s)
| | - Majid Jabir
- Department of Applied Sciences, University of Technology, Iraq
| | | | - Malathi Hanumanthaiah
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Neha Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri - 140307, Mohali, Punjab, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University Dehradun, Uttarakhand, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Sabrean Farhan Jawad
- Department of Biochemistry, College of Science, Al-Mustaqbal University, 51001 Babil, Iraq.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Delta State University, Faculty of Pharmacy, PMB 1 Abraka, Delta State, Nigeria
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
31
|
Dai Y, Tian X, Ye X, Gong Y, Xu L, Jiao L. Role of the TME in immune checkpoint blockade resistance of non-small cell lung cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:52. [PMID: 39802954 PMCID: PMC11724356 DOI: 10.20517/cdr.2024.166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025]
Abstract
Primary and secondary resistance to immune checkpoint blockade (ICB) reduces its efficacy. The mechanisms underlying immunotherapy resistance are highly complex. In non-small cell lung cancer (NSCLC), these mechanisms are primarily associated with the loss of programmed cell death-ligand 1 (PD-L1) expression, genetic mutations, circular RNA axis and transcription factor regulation, antigen presentation disorders, and dysregulation of signaling pathways. Additionally, alterations in the tumor microenvironment (TME) play a pivotal role in driving immunotherapy resistance. Primary resistance is mainly attributed to TME alterations, including mutations and co-mutations, modulation of T cell infiltration, enrichment of M2 tumor-associated macrophages (M2-TAMs) and mucosal-associated invariant T (MAIT) cells, vascular endothelial growth factor (VEGF), and pulmonary fibrosis. Acquired resistance mainly stems from changes in cellular infiltration patterns leading to "cold" or "hot" tumors, altered interferon (IFN) signaling pathway expression, involvement of extracellular vesicles (EVs), and oxidative stress responses, as well as post-treatment gene mutations and circadian rhythm disruption (CRD). This review presents an overview of various mechanisms underlying resistance to ICB, elucidates the alterations in the TME during primary, adaptive, and acquired resistance, and discusses existing strategies for overcoming ICB resistance.
Collapse
Affiliation(s)
- Yuening Dai
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xueqi Tian
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xuanting Ye
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yabin Gong
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Ling Xu
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Translational Cancer Research for Integrated Chinese and Western Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Lijing Jiao
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Translational Cancer Research for Integrated Chinese and Western Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| |
Collapse
|
32
|
Weller J, Potthoff A, Zeyen T, Schaub C, Duffy C, Schneider M, Herrlinger U. Current status of precision oncology in adult glioblastoma. Mol Oncol 2024; 18:2927-2950. [PMID: 38899374 PMCID: PMC11619805 DOI: 10.1002/1878-0261.13678] [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: 11/16/2023] [Revised: 04/05/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The concept of precision oncology, the application of targeted drugs based on comprehensive molecular profiling, has revolutionized treatment strategies in oncology. This review summarizes the current status of precision oncology in glioblastoma (GBM), the most common and aggressive primary brain tumor in adults with a median survival below 2 years. Targeted treatments without prior target verification have consistently failed. Patients with BRAF V600E-mutated GBM benefit from BRAF/MEK-inhibition, whereas targeting EGFR alterations was unsuccessful due to poor tumor penetration, tumor cell heterogeneity, and pathway redundancies. Systematic screening for actionable molecular alterations resulted in low rates (< 10%) of targeted treatments. Efficacy was observed in one-third and currently appears to be limited to BRAF-, VEGFR-, and mTOR-directed treatments. Advancing precision oncology for GBM requires consideration of pathways instead of single alterations, new trial concepts enabling rapid and adaptive drug evaluation, a focus on drugs with sufficient bioavailability in the CNS, and the extension of target discovery and validation to the tumor microenvironment, tumor cell networks, and their interaction with immune cells and neurons.
Collapse
Affiliation(s)
- Johannes Weller
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | | | - Thomas Zeyen
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | - Christina Schaub
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | - Cathrina Duffy
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| | | | - Ulrich Herrlinger
- Department of Neurooncology, Center for NeurologyUniversity Hospital BonnGermany
| |
Collapse
|
33
|
Gao H, Zhu J, Wu T, Long Q, Guan X, Chen Q, Yi W. Comprehensive pancancer analysis reveals that LPCAT1 is a novel predictive biomarker for prognosis and immunotherapy response. Apoptosis 2024; 29:2128-2146. [PMID: 39097858 DOI: 10.1007/s10495-024-02010-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/05/2024]
Abstract
Lysophosphatidylcholine acyltransferase 1 (LPCAT1) is a crucial enzyme involved in phospholipid metabolism and is essential for maintaining the structure and functionality of biofilms. However, a comprehensive examination of the role of LPCAT1 across various cancer types is lacking. Multiple public databases have been utilized to examine LPCAT1 expression, genetic alterations, methylation, prognosis, biological function, and its relationship with antitumor immunity in different cancer types. The function of LPCAT1 in glioma, breast cancer and liver cancer cells was further verified using in vitro experiments. Our research indicated that LPCAT1 is upregulated in various cancers and is accompanied by a wide range of amplification mutations. Higher LPCAT1 expression was associated with poorer prognosis across multiple cancers. Further in vitro experiments demonstrated that interfering with LPCAT1 expression increased apoptosis in glioma, breast cancer and liver cancer cells and concurrently suppressed their proliferation and migration. Functional enrichment analysis revealed that LPCAT1-associated genes were primarily enriched in immune and cancer progression pathways, such as the JAK/STAT, MYC, and EMT, etc. Moreover, LPCAT1 expression was closely associated with immune cell infiltration and immune checkpoint-related gene expression. Interestingly, LPCAT1 expression levels were generally higher in patients in the immunotherapy response group. The combination of LPCAT1 and PDL1 serves as an effective predictor of immunotherapy response. In conclusion, LPCAT1 is involved in immune regulation and tumor progression and holds promise as a biomarker for predicting patient outcomes and immunotherapy efficacy.
Collapse
Affiliation(s)
- Hongyu Gao
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Jinfeng Zhu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Tong Wu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Qian Long
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Xinyu Guan
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Qitong Chen
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan, China.
| |
Collapse
|
34
|
Han H, Li Z, Feng Y, Song H, Fang Z, Zhang D, Yuan D, Shi J. Peptide Degrader-Based Targeting of METTL3/14 Improves Immunotherapy Response in Cutaneous Melanoma. Angew Chem Int Ed Engl 2024; 63:e202407381. [PMID: 39136347 DOI: 10.1002/anie.202407381] [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: 04/18/2024] [Indexed: 10/30/2024]
Abstract
METTL3 has emerged as a promising therapeutic target in cancer treatment, although its oncogenic functions in melanoma development and potential for therapeutic targeting drug have not been fully explored. In this study, we define the oncogenic role of METTL3 in melanoma development and progression. Building on this insight, we examine our recently designed peptide inhibitor RM3, which targets the binding interface of METTL3/14 complex for disruption and subsequent ubiquitin-mediated proteasomal degradation via the E3 ligase STUB1. RM3 treatment reduces proliferation, migration, and invasion, and induces apoptosis in melanoma cells in vitro and in vivo. Subsequent transcriptomic analysis identified changes in immuno-related genes following RM3-mediated suppression of METTL3/14 N6-methyladenosine (m6A) methyltransferase activity, suggesting a potential for interaction with immunotherapy. A combination treatment of RM3 with anti-PD-1 antibody results in significantly higher beneficial tumor response in vivo, with a good safety profile. Collectively, these findings not only delineate the oncogenic role of METTL3 in melanoma but also showcase RM3, acting as a peptide degrader, as a novel and promising strategy for melanoma treatment.
Collapse
Affiliation(s)
- Hong Han
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, P. R. China
| | - Zenghui Li
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, P. R. China
| | - Yuqing Feng
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, P. R. China
| | - He Song
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, P. R. China
| | - Zhixiong Fang
- Affiliated Hospital of Hunan University, Department of Infectious Disease and Public Health, Hunan province, P. R. China
| | - Dingxiao Zhang
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, P. R. China
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, 518000, P. R. China
| | - Dan Yuan
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, P. R. China
| | - Junfeng Shi
- Affiliated Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha, P. R. China
- Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, 518000, P. R. China
| |
Collapse
|
35
|
Fujimura T, Furugaki K, Mizuta H, Muraoka S, Nishio M, Adachi J, Uchibori K, Miyauchi E, Hayashi H, Katayama R, Yoshiura S. Targeting ErbB and tankyrase1/2 prevent the emergence of drug-tolerant persister cells in ALK-positive lung cancer. NPJ Precis Oncol 2024; 8:264. [PMID: 39551860 PMCID: PMC11570601 DOI: 10.1038/s41698-024-00757-w] [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: 09/07/2023] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
Targeting the drug tolerant persister (DTP) state in cancer cells should prevent further development of resistance mechanisms. This study explored combination therapies to inhibit alectinib-induced DTP cell formation from anaplastic lymphoma kinase-positive non-small cell lung cancer (ALK + NSCLC) patient-derived cells. After drug-screening 3114 compounds, pan-HER inhibitors (ErbB pathway) and tankyrase1/2 inhibitors (Wnt/β-catenin signaling) emerged as top candidates to inhibit alectinib-induced DTP cells growth. We confirmed knockdown of both TNKS1/2 in DTP cells recovered the sensitivity to alectinib. Further, our study suggested knockdown of TNKS1/2 increased stability of Axin1/2, which induced β-catenin degradation and decreased its nuclear translocation, thereby suppressing transcription of antiapoptotic and proliferation-related genes (survivin, c-MYC). Targeting both pathways with alectinib+pan-HER inhibitor and alectinib+TNKS1/2 inhibitor suppressed alectinib-induced DTP cells, and the triple combination almost completely prevented the appearance of DTP cells. In conclusion, combination with ALK-TKI, pan-HER and TNKS1/2 inhibitors has the potential to prevent the emergence of DTP in ALK + NSCLC.
Collapse
Affiliation(s)
- Takaaki Fujimura
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Yokohama, Japan
| | - Koh Furugaki
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Yokohama, Japan
| | - Hayato Mizuta
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Yokohama, Japan
| | - Satoshi Muraoka
- Laboratory of Proteomics for Drug Discovery, Laboratory of Clinical and Analytical Chemistry, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Makoto Nishio
- Department of Respiratory Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Laboratory of Clinical and Analytical Chemistry, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Ken Uchibori
- Department of Respiratory Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Eisaku Miyauchi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Sayama, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Shigeki Yoshiura
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Yokohama, Japan.
| |
Collapse
|
36
|
Ji H, Ma W, Zheng A, Tang D. The role and molecular mechanism of Trametes Robiniophila Murr(Huaier) in tumor therapy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118578. [PMID: 39004194 DOI: 10.1016/j.jep.2024.118578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/12/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Trametes Robiniophila Murr, commonly known as Huaier, has been extensively documented in ethnopharmacology research in China. Huaier has a long history of clinical usage spanning over 1000 years in China. Traditional clinical application records demonstrate the wide utilization of Huaier for treating various cancers and enhancing the autoimmunity of tumor patients. AIM OF THE REVIEW The present study provides a comprehensive review of the traditional uses, phytochemical constituents, pharmacological activities, anti-tumor mechanism, and potential applications of Huaier, thereby offering valuable insights for the further development and utilization of this natural product. MATERIALS AND METHODS This study employed the keywords "Trametes Robiniophila Murr" and "Huaier" to retrieve relevant information on Huaier from various databases, including PubMed, Web of Science, Springer, Science Direct, ACS, Wiley, CNKI, Baidu Scholar, Google Scholar, and ancient materia medica. RESULTS Trametes Robiniophila Murr (Huaier), a traditional Chinese medicine, has demonstrated significant efficacy in the clinical treatment of various tumors. The primary bioactive constituents of Huaier consist of fungal-derived compounds, including polysaccharides, proteins, ketones, alkaloids, and minerals. The research findings demonstrate that Huaier serves as a reliable adjunctive therapeutic agent by effectively inhibiting cancer cell proliferation, inducing apoptosis in cancer cells, suppressing tumor metastasis, regulating tumor stem cells and immune function. Therefore, it exerts a potent anti-tumor effect when used in conjunction with conventional anti-cancer therapies. CONCLUSIONS The analysis of traditional uses, phytochemical composition, and pharmacological activity reveals that Huaier exhibits significant potential as a medicinal plant with diverse pharmacological effects. Owing to its numerous advantages, Huaier holds immense promise for application in the domains of tumor prevention and treatment, enhancing both survival time and quality of life among cancer patients.
Collapse
Affiliation(s)
- Hao Ji
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225000, China.
| | - Wei Ma
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225000, China.
| | - Aiyu Zheng
- Department of Geriatrics, Taixing People's Hospital, Taixing, 225400, China.
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital; Northern Jiangsu People's Hospital Affiliated to Yangzhou University; The Yangzhou Clinical Medical College of Xuzhou Medical University; The Yangzhou School of Clinical Medicine of Dalian Medical University; The Yangzhou School of Clinical Medicine of Nanjing Medical University; Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, 225000, China.
| |
Collapse
|
37
|
Xiang X, Wang K, Zhang H, Mou H, Shi Z, Tao Y, Song H, Lian Z, Wang S, Lu D, Wei X, Xie H, Zheng S, Wang J, Xu X. Blocking CX3CR1+ Tumor-Associated Macrophages Enhances the Efficacy of Anti-PD1 Therapy in Hepatocellular Carcinoma. Cancer Immunol Res 2024; 12:1603-1620. [PMID: 39115356 DOI: 10.1158/2326-6066.cir-23-0627] [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: 08/01/2023] [Revised: 04/08/2024] [Accepted: 08/07/2024] [Indexed: 11/05/2024]
Abstract
The efficacy of immune checkpoint inhibitors in the treatment of hepatocellular carcinoma (HCC) remains limited, highlighting the need for further investigation into the mechanisms underlying treatment resistance. Accumulating evidence indicates that tumor-associated macrophages (TAM) within the tumor microenvironment demonstrate a key role in immune evasion and treatment resistance. This study explored the role of TAMs in the HCC tumor microenvironment. Our findings reveal that TAMs expressing CX3C motif chemokine receptor 1 (CX3CR1) induced T-cell exhaustion through IL27 secretion in orthotopic models of HCC following treatment with anti-PD1. Moreover, we identified prostaglandin E2 (PGE2), released by immune-attacked tumor cells, as a key regulator of TAM transition to a CX3CR1+ phenotype. To augment the therapeutic response to anti-PD1 therapy, we propose targeting CX3CR1+ TAMs in addition to anti-PD1 therapy. Our study contributes to the understanding of the role of TAMs in cancer immunotherapy and highlights potential clinical implications for HCC treatment. The combination of targeting CX3CR1+ TAMs with anti-PD1 therapy holds promise for enhancing the efficacy of immunotherapeutic interventions in patients with HCC.
Collapse
Affiliation(s)
- Xiaonan Xiang
- Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Wang
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Hui Zhang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Haibo Mou
- Department of Medical Oncology, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Zhixiong Shi
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yaoye Tao
- Zhejiang University School of Medicine, Hangzhou, China
| | - Hongliang Song
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Zhengxing Lian
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Di Lu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Xuyong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Haiyang Xie
- Department of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Shusen Zheng
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Department of Hepatobiliary & Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
38
|
Vijayan Y, James S, Viswanathan A, Aparna JS, Bindu A, Namitha NN, Anantharaman D, Babu Lankadasari M, Harikumar KB. Targeting acid ceramidase enhances antitumor immune response in colorectal cancer. J Adv Res 2024; 65:73-87. [PMID: 38142035 PMCID: PMC11518951 DOI: 10.1016/j.jare.2023.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 12/25/2023] Open
Abstract
INTRODUCTION Acid ceramidase (hereafter referred as ASAH1) is an enzyme in sphingolipid metabolism that converts pro-survival ceramide into sphingosine. ASAH1 has been shown to be overexpressed in certain cancers. However, the role of ASAH1 in colorectal cancer still remain elusive. OBJECTIVE The present study is aimed to understand how ASAH1 regulates colorectal cancer (CRC) progression and resistance to checkpoint inhibitor therapy. METHODS Both pharmacological and genetic silencing of ASAH1 was used in the study. In vitro experiments were done on human and mouse CRC cell lines. The in vivo studies were conducted in NOD-SCID and BALB/c mice models. The combination of ASAH1 inhibitor and checkpoint inhibitor was tested using a syngeneic tumor model of CRC. Transcriptomic and metabolomic analyses were done to understand the effect of ASAH1 silencing. RESULTS ASAH1 is overexpressed in human CRC cases, and silencing the expression resulted in the induction of immunological cell death (ICD) and mitochondrial stress. The ASAH1 inhibitor (LCL-521), either as monotherapy or in combination with an anti-PD-1 antibody, resulted in reduction of tumors and, through induction of type I and II interferon response, activation of M1 macrophages and T cells, leading to enhanced infiltration of cytotoxic T cells. Our findings supported that the combination of LCL-521 and ICIs, which enhances the antitumor responses, and ASAH1 can be a druggable target in CRC.
Collapse
Affiliation(s)
- Yadu Vijayan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Shirley James
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Arun Viswanathan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India; Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Jayasekharan S Aparna
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Anu Bindu
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Narayanan N Namitha
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Devasena Anantharaman
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Manendra Babu Lankadasari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, India.
| |
Collapse
|
39
|
Liu S, Zhang L, Ding K, Zeng B, Li B, Zhou J, Li J, Wang J, Zhang H, Sun R, Su X. S. glabra exerts anti-lung cancer effects by inducing ferroptosis and anticancer immunity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155981. [PMID: 39260134 DOI: 10.1016/j.phymed.2024.155981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/09/2024] [Accepted: 08/22/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Sarcandra glabra (S. glabra), a traditional Chinese medicine (TCM), has demonstrated significant anticancer activity; however, the underlying mechanisms have not yet been fully elucidated. PURPOSE This study aimed to investigate the effects of S. glabra on lung cancer and to explore its underlying mechanisms. METHODS The chemical profile of S. glabra was analyzed via ultrahigh-performance liquid chromatography coupled with mass spectrometry (UPLC-MS). The effects of S. glabra on the viability, proliferation, apoptosis, migration, and invasion of lung cancer cells were assessed via CCK8, colony formation, flow cytometry, scratch, and Transwell assays. In vivo anticancer activity was evaluated in an LLC mouse model. Proteomic analysis was performed to identify key molecules and pathways in S. glabra-treated LLC cells. The expression of ferroptotic proteins and associated cellular events were examined via western blotting, ROS production, iron accumulation, and lipid peroxidation assays. Immune modulation in tumor-bearing mice was evaluated by detecting immune cells and cytokines in the peripheral blood and tumor tissue. RESULTS Our analysis quantified 1997 chemical markers in S. glabra aqueous extracts. S. glabra inhibited the viability and proliferation of lung cancer cells and induced cell cycle arrest and apoptosis. Scratch and Transwell assays demonstrated that S. glabra suppressed the migration and invasion of lung cancer cells. Oral administration of S. glabra significantly inhibited tumor growth in LLC tumor-bearing mice. Proteomic analysis revealed that S. glabra upregulated the expression of the HMOX1 protein and activated the ferroptosis pathway. Consistent with these findings, we found that S. glabra triggered ferroptosis in lung cancer cells, as evidenced by the upregulation of HMOX1, downregulation of GPX4 and ferritin light chain proteins, iron accumulation, increased ROS production, and lipid peroxidation. Furthermore, S. glabra demonstrated immunostimulatory properties in LLC tumor-bearing mice, leading to increased populations of immune cells (NK cells) and elevated cytokine levels (IL-2). CONCLUSION This study is the first to demonstrate that S. glabra induces ferroptosis in lung cancer cells by regulating HMOX1, GPX4, and FTL. These findings provide a robust scientific basis for the clinical application of S. glabra in lung cancer treatment.
Collapse
Affiliation(s)
- Songyu Liu
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Lu Zhang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Kai Ding
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Bin Zeng
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Bo Li
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Jinyi Zhou
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Jv Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Junliang Wang
- Scientific Research and Experimental Center, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Huijun Zhang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai 200040, China.
| | - Ruifen Sun
- School of Nursing, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Xiaosan Su
- Scientific Research and Experimental Center, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
40
|
Chen Q, Zheng X, Cheng W, Li J. Landscape of targeted therapies for lung squamous cell carcinoma. Front Oncol 2024; 14:1467898. [PMID: 39544292 PMCID: PMC11560903 DOI: 10.3389/fonc.2024.1467898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
Lung cancer, a common type of malignant neoplasm, has seen significant advancements in the treatment of lung adenocarcinoma (LUAD). However, the management of lung squamous cell carcinoma (LSCC) continues to pose challenges. Traditional treatment methods for LSCC encompass surgical resection, chemotherapy, and radiotherapy. The introduction of targeted therapy and immunotherapy has greatly benefited LSCC patients, but issues such as limited immune response rates and adverse reactions persist. Therefore, gaining a deeper comprehension of the underlying mechanisms holds immense importance. This review provides an in-depth overview of classical signaling pathways and therapeutic targets, including the PI3K signaling pathway, CDK4/6 pathway, FGFR1 pathway and EGFR pathway. Additionally, we delve into alternative signaling pathways and potential targets that could offer new therapeutic avenues for LSCC. Lastly, we summarize the latest advancements in targeted therapy combined with immune checkpoint blockade (ICB) therapy for LSCC and discuss the prospects and challenges in this field.
Collapse
Affiliation(s)
- Qiuxuan Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoshuo Zheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weiting Cheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jian Li
- Institude of Experimental Immunology, University Clinic of Rheinische Friedrich-Wihelms-University, Bonn, Germany
| |
Collapse
|
41
|
Rizzo A, Brunetti O, Brandi G. Hepatocellular Carcinoma Immunotherapy: Predictors of Response, Issues, and Challenges. Int J Mol Sci 2024; 25:11091. [PMID: 39456872 PMCID: PMC11507510 DOI: 10.3390/ijms252011091] [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: 08/24/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs), such as durvalumab, tremelimumab, and atezolizumab, have emerged as a significant therapeutic option for the treatment of hepatocellular carcinoma (HCC). In fact, the efficacy of ICIs as single agents or as part of combination therapies has been demonstrated in practice-changing phase III clinical trials. However, ICIs confront several difficulties, including the lack of predictive biomarkers, primary and secondary drug resistance, and treatment-related side effects. Herein, we provide an overview of current issues and future challenges in this setting.
Collapse
Affiliation(s)
- Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Oronzo Brunetti
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Giovanni Brandi
- Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Via Giuseppe Massarenti, 9, 40138 Bologna, Italy
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni, 15, 40138 Bologna, Italy
| |
Collapse
|
42
|
Vaena SG, Romeo MJ, Mina-Abouda M, Funk EC, Fullbright G, Long DT, Delaney JR. Autophagy unrelated transcriptional mechanisms of hydroxychloroquine resistance revealed by integrated multi-omics of evolved cancer cells. Cell Cycle 2024:1-21. [PMID: 39299930 DOI: 10.1080/15384101.2024.2402191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/01/2024] [Accepted: 07/25/2024] [Indexed: 09/22/2024] Open
Abstract
Hydroxychloroquine (HCQ) and chloroquine are repurposed drugs known to disrupt autophagy, a molecular recycling pathway essential for tumor cell survival, chemotherapeutic resistance, and stemness. We pursued a multi-omic strategy in OVCAR3 ovarian cancer and CCL218 colorectal cancer cells. Two genome-scale screens were performed. In the forward genetic screen, cell populations were passaged for 15 drug pulse-chases with HCQ or vehicle control. Evolved cells were collected and processed for bulk RNA-seq, exome-seq, and single-cell RNA-seq (scRNA-seq). In the reverse genetic screen, a pooled CRISPR-Cas9 library was used in cells over three pulse-chases of HCQ or vehicle control treatments. HCQ evolved cells displayed remarkably few mutational differences, but substantial transcriptional differences. Transcriptomes revealed multiple pathways associated with resistance to HCQ, including upregulation of glycolysis, exocytosis, and chromosome condensation/segregation, or downregulation of translation and apoptosis. The Cas9 screen identified only one autophagy gene. Chromosome condensation and segregation were confirmed to be disrupted by HCQ in live cells and organelle-free in vitro extracts. Transcriptional plasticity was the primary mechanism by which cells evolved resistance to HCQ. Neither autophagy nor the lysosome were substantive hits. Our analysis may serve as a model for how to better position repurposed drugs in oncology.
Collapse
Affiliation(s)
- Silvia G Vaena
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Martin J Romeo
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Mirna Mina-Abouda
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Emma C Funk
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - George Fullbright
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - David T Long
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Joe R Delaney
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
43
|
Lin Z, Li G, Jiang K, Li Z, Liu T. Cancer therapy resistance mediated by cancer-associated fibroblast-derived extracellular vesicles: biological mechanisms to clinical significance and implications. Mol Cancer 2024; 23:191. [PMID: 39244548 PMCID: PMC11380334 DOI: 10.1186/s12943-024-02106-8] [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: 07/31/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a diverse stromal cell population within the tumour microenvironment, where they play fundamental roles in cancer progression and patient prognosis. Multiple lines of evidence have identified that CAFs are critically involved in shaping the structure and function of the tumour microenvironment with numerous functions in regulating tumour behaviours, such as metastasis, invasion, and epithelial-mesenchymal transition (EMT). CAFs can interact extensively with cancer cells by producing extracellular vesicles (EVs), multiple secreted factors, and metabolites. Notably, CAF-derived EVs have been identified as critical mediators of cancer therapy resistance, and constitute novel therapy targets and biomarkers in cancer management. This review aimed to summarize the biological roles and detailed molecular mechanisms of CAF-derived EVs in mediating cancer resistance to chemotherapy, targeted therapy agents, radiotherapy, and immunotherapy. We also discussed the therapeutic potential of CAF-derived EVs as novel targets and clinical biomarkers in cancer clinical management, thereby providing a novel therapeutic strategy for enhancing cancer therapy efficacy and improving patient prognosis.
Collapse
Affiliation(s)
- Zhengjun Lin
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139# Middle Renmin Road, Changsha, Hunan Province, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Guoqing Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139# Middle Renmin Road, Changsha, Hunan Province, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Ke Jiang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139# Middle Renmin Road, Changsha, Hunan Province, 410011, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139# Middle Renmin Road, Changsha, Hunan Province, 410011, China.
| | - Tang Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139# Middle Renmin Road, Changsha, Hunan Province, 410011, China.
| |
Collapse
|
44
|
Wang L, Lin Y, Yao Z, Babu N, Lin W, Chen C, Du L, Cai S, Pan Y, Xiong X, Ye Q, Ren H, Zhang D, Chen Y, Yeung SCJ, Bremer E, Zhang H. Targeting undruggable phosphatase overcomes trastuzumab resistance by inhibiting multi-oncogenic kinases. Drug Resist Updat 2024; 76:101118. [PMID: 39094301 DOI: 10.1016/j.drup.2024.101118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/12/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024]
Abstract
AIMS Resistance to targeted therapy is one of the critical obstacles in cancer management. Resistance to trastuzumab frequently develops in the treatment for HER2+ cancers. The role of protein tyrosine phosphatases (PTPs) in trastuzumab resistance is not well understood. In this study, we aim to identify pivotal PTPs affecting trastuzumab resistance and devise a novel counteracting strategy. METHODS Four public datasets were used to screen PTP candidates in relation to trastuzumab responsiveness in HER2+ breast cancer. Tyrosine kinase (TK) arrays were used to identify kinases that linked to protein tyrosine phosphate receptor type O (PTPRO)-enhanced trastuzumab sensitivity. The efficacy of small activating RNA (saRNA) in trastuzumab-conjugated silica nanoparticles was tested for PTPRO upregulation and resistance mitigation in cell models, a transgenic mouse model, and human cancer cell line-derived xenograft models. RESULTS PTPRO was identified as the key PTP which influences trastuzumab responsiveness and patient survival. PTPRO de-phosphorated several TKs, including the previously overlooked substrate ERBB3, thereby inhibiting multiple oncogenic pathways associated with drug resistance. Notably, PTPRO, previously deemed "undruggable," was effectively upregulated by saRNA-loaded nanoparticles. The upregulated PTPRO simultaneously inhibited ERBB3, ERBB2, and downstream SRC signaling pathways, thereby counteracting trastuzumab resistance. CONCLUSIONS Antibody-conjugated saRNA represents an innovative approach for targeting "undruggable" PTPs.
Collapse
Affiliation(s)
- Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Yusheng Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China; Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; Department of Hematology, University of Groningen, University Medical Center Groningen, the Netherlands; Shantou University Medical College, Shantou, China
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China; Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Nipun Babu
- Shantou University Medical College, Shantou, China
| | - Wan Lin
- Shantou University Medical College, Shantou, China
| | | | - Liang Du
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Songwang Cai
- Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yunlong Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao Xiong
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Qiantao Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Hongzheng Ren
- Department of Pathology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China; Department of Pathology, Heping Hospital, Changzhi Medical College, Changzhi, China
| | - Dianzheng Zhang
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Yexi Chen
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin Bremer
- Department of Hematology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Hao Zhang
- Department of Pathology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
45
|
Ou X, Gao G, Habaz IA, Wang Y. Mechanisms of resistance to tyrosine kinase inhibitor-targeted therapy and overcoming strategies. MedComm (Beijing) 2024; 5:e694. [PMID: 39184861 PMCID: PMC11344283 DOI: 10.1002/mco2.694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
Tyrosine kinase inhibitor (TKI)-targeted therapy has revolutionized cancer treatment by selectively blocking specific signaling pathways crucial for tumor growth, offering improved outcomes with fewer side effects compared with conventional chemotherapy. However, despite their initial effectiveness, resistance to TKIs remains a significant challenge in clinical practice. Understanding the mechanisms underlying TKI resistance is paramount for improving patient outcomes and developing more effective treatment strategies. In this review, we explored various mechanisms contributing to TKI resistance, including on-target mechanisms and off-target mechanisms, as well as changes in the tumor histology and tumor microenvironment (intrinsic mechanisms). Additionally, we summarized current therapeutic approaches aiming at circumventing TKI resistance, including the development of next-generation TKIs and combination therapies. We also discussed emerging strategies such as the use of dual-targeted antibodies and PROteolysis Targeting Chimeras. Furthermore, we explored future directions in TKI-targeted therapy, including the methods for detecting and monitoring drug resistance during treatment, identification of novel targets, exploration of dual-acting kinase inhibitors, application of nanotechnologies in targeted therapy, and so on. Overall, this review provides a comprehensive overview of the challenges and opportunities in TKI-targeted therapy, aiming to advance our understanding of resistance mechanisms and guide the development of more effective therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
- Xuejin Ou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Ge Gao
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China HospitalSichuan UniversityChengduChina
| | - Inbar A. Habaz
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
46
|
Wang S, Guo S, Guo J, Du Q, Wu C, Wu Y, Zhang Y. Cell death pathways: molecular mechanisms and therapeutic targets for cancer. MedComm (Beijing) 2024; 5:e693. [PMID: 39239068 PMCID: PMC11374700 DOI: 10.1002/mco2.693] [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: 04/08/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 09/07/2024] Open
Abstract
Cell death regulation is essential for tissue homeostasis and its dysregulation often underlies cancer development. Understanding the different pathways of cell death can provide novel therapeutic strategies for battling cancer. This review explores several key cell death mechanisms of apoptosis, necroptosis, autophagic cell death, ferroptosis, and pyroptosis. The research gap addressed involves a thorough analysis of how these cell death pathways can be precisely targeted for cancer therapy, considering tumor heterogeneity and adaptation. It delves into genetic and epigenetic factors and signaling cascades like the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathways, which are critical for the regulation of cell death. Additionally, the interaction of the microenvironment with tumor cells, and particularly the influence of hypoxia, nutrient deprivation, and immune cellular interactions, are explored. Emphasizing therapeutic strategies, this review highlights emerging modulators and inducers such as B cell lymphoma 2 (BCL2) homology domain 3 (BH3) mimetics, tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), chloroquine, and innovative approaches to induce ferroptosis and pyroptosis. This review provides insights into cancer therapy's future direction, focusing on multifaceted approaches to influence cell death pathways and circumvent drug resistance. This examination of evolving strategies underlines the considerable clinical potential and the continuous necessity for in-depth exploration within this scientific domain.
Collapse
Affiliation(s)
- Shaohui Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Sa Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Jing Guo
- College of Clinical Medicine Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Qinyun Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Cen Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Yeke Wu
- College of Clinical Medicine Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Yi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine Chengdu University of Traditional Chinese Medicine Chengdu China
| |
Collapse
|
47
|
Hong Z, Liu F, Zhang Z. Ubiquitin modification in the regulation of tumor immunotherapy resistance mechanisms and potential therapeutic targets. Exp Hematol Oncol 2024; 13:91. [PMID: 39223632 PMCID: PMC11367865 DOI: 10.1186/s40164-024-00552-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Although immune checkpoint-based cancer immunotherapy has shown significant efficacy in various cancers, resistance still limits its therapeutic effects. Ubiquitination modification is a mechanism that adds different types of ubiquitin chains to proteins, mediating protein degradation or altering their function, thereby affecting cellular signal transduction. Increasing evidence suggests that ubiquitination modification plays a crucial role in regulating the mechanisms of resistance to cancer immunotherapy. Drugs targeting ubiquitination modification pathways have been shown to inhibit tumor progression or enhance the efficacy of cancer immunotherapy. This review elaborates on the mechanisms by which tumor cells, immune cells, and the tumor microenvironment mediate resistance to cancer immunotherapy and the details of how ubiquitination modification regulates these mechanisms, providing a foundation for enhancing the efficacy of cancer immunotherapy by intervening in ubiquitination modification.
Collapse
Affiliation(s)
- Zihang Hong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China
| | - Furong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
48
|
Thakur N, Singh P, Bagri A, Srivastava S, Dwivedi V, Singh A, Jaiswal SK, Dholpuria S. Therapy resistance in prostate cancer: mechanism, signaling and reversal strategies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1110-1134. [PMID: 39351434 PMCID: PMC11438573 DOI: 10.37349/etat.2024.00266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024] Open
Abstract
Prostate cancer (PC) depicts a major health challenge all over the globe due to its complexities in the treatment and diverse clinical trajectories. Even in the advances in the modern treatment strategies, the spectrum of resistance to the therapies continues to be a significant challenge. This review comprehensively examines the underlying mechanisms of the therapy resistance occurred in PC, focusing on both the tumor microenvironment and the signaling pathways implicated in the resistance. Tumor microenvironment comprises of stromal and epithelial cells, which influences tumor growth, response to therapy and progression. Mechanisms such as microenvironmental epithelial-mesenchymal transition (EMT), anoikis suppression and stimulation of angiogenesis results in therapy resistance. Moreover, dysregulation of signaling pathways including androgen receptor (AR), mammalian target of rapamycin/phosphoinositide 3 kinase/AKT (mTOR/PI3K/AKT), DNA damage repair and Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways drive therapy resistance by promoting tumor survival and proliferation. Understanding these molecular pathways is important for developing targeted therapeutic interventions which overcomes resistance. In conclusion, a complete grasp of mechanisms and pathways underlying medication resistance in PC is important for the development of individualized treatment plans and enhancements of clinical outcomes. By studying and understanding the complex mechanisms of signaling pathways and microenvironmental factors contributing to therapy resistance, this study focuses and aims to guide the development of innovative therapeutic approaches to effectively overcome the PC progression and improve the survival rate of patients.
Collapse
Affiliation(s)
- Neha Thakur
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Pallavi Singh
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Aditi Bagri
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Saumya Srivastava
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand 248002, India
| | - Vinay Dwivedi
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh 474005, India
| | - Asha Singh
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh 474005, India
| | - Sunil Kumar Jaiswal
- School of Biological and Life Sciences, Galgotias University, Greater Noida, Uttar Pradesh 203201, India
| | - Sunny Dholpuria
- Department of Life Sciences, J. C. Bose University of Science and Technology, YMCA Faridabad, Faridabad, Haryana 121006, India
| |
Collapse
|
49
|
Sood A, Jothiswaran V, Singh A, Sharma A. Anticancer peptides as novel immunomodulatory therapeutic candidates for cancer treatment. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1074-1099. [PMID: 39351437 PMCID: PMC11438574 DOI: 10.37349/etat.2024.00264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/27/2024] [Indexed: 10/04/2024] Open
Abstract
Cancer remains a concern after years of research in this field. Conventional therapies such as chemotherapy, radiation, and surgery are available for cancer treatment, but they are characterized by various side effects. There are several immunological challenges that make it difficult for the immune system and conventional therapies to treat cancer. Some of these challenges include heterogeneity, resistance to medicines, and cancer relapse. Even advanced treatments like immune checkpoint inhibitors (ICIs), which revolutionized cancer treatment, have associated toxicity and resistance further necessitate the exploration of alternative therapies. Anticancer peptides (ACPs) offer promising potential as cancer-fighting agents and address challenges such as treatment resistance, tumor heterogeneity, and metastasis. Although these peptides exist as components of the defense system in various plants, animals, fungi, etc., but can also be created synthetically and used as a new treatment measure. These peptides possess properties that make them appealing for cancer therapy, such as apoptosis induction, inhibition of angiogenesis, and cell membrane breakdown with low toxicity. Their capacity to specifically target cancer cells selectively holds promise for enhancing treatment environments as well as improving patients' quality of life. This review provides detailed insights into the different prospects of ACPs, including their characterization, use as immunomodulatory agents in cancer treatment, and their mechanistic details after addressing various immunological challenges in existing cancer treatment strategies. In conclusion, ACPs have promising potential as novel cancer therapeutics due to their target specificity and fewer side effects than conventional therapies.
Collapse
Affiliation(s)
- Apurva Sood
- Department of Molecular Biology and Genetic Engineering, School of Bioengineering and Biosciences, Lovely Professional University, Punjab 144411, India
| | - V.V. Jothiswaran
- Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769005, India
| | - Amrita Singh
- Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769005, India
| | - Anuradha Sharma
- Department of Molecular Biology and Genetic Engineering, School of Bioengineering and Biosciences, Lovely Professional University, Punjab 144411, India
| |
Collapse
|
50
|
Huang X, Zhang W. Overcoming T Cell Exhaustion in Tumor Microenvironment via Immune Checkpoint Modulation with Nano-Delivery Systems for Enhanced Immunotherapy. SMALL METHODS 2024; 8:e2301326. [PMID: 38040834 DOI: 10.1002/smtd.202301326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/12/2023] [Indexed: 12/03/2023]
Abstract
Immune checkpoint blockade (ICB) therapy for tumors has arisen in growing interest. However, the low response rate of tumors to ICB is mainly attributed to the inhibitory infiltration of immune cells in the tumor microenvironment (TME). Despite the promising benefits of ICB, the therapeutic effects of antibodies are dependent on a high dose and long-term usage in the clinic, thereby leading to immune-related adverse effects. Accordingly, ICB combined with nano-delivery systems could be used to overcome T cell exhaustion, which reduces the side effects and the usage of antibodies with higher response rates in patients. In this review, the authors aim to overcome T cell exhaustion in TME via immune checkpoint modulation with nano-delivery systems for enhanced immunotherapy. Several strategies are summarized to combine ICB and nano-delivery systems to further enhance immunotherapy: a) expressing immune checkpoint on the surface of nano-delivery systems; b) loading immune checkpoint inhibitors into nano-delivery systems; c) loading gene-editing technology into nano-delivery systems; and d) nano-delivery systems mediated immune checkpoint modulation. Taken together, ICB combined with nano-delivery systems might be a promising strategy to overcome T cell exhaustion in TME for enhanced immunotherapy.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|