1
|
Manikandan DB, Jagadeeshan S, Mathukkada S, Shareb RA, Prasad M, Belsamma LVS, Marripati D, Erez N, Wainer M, Geva A, Raviv D, Allon I, Morris LG, Su GH, Wang H, Rosenberg AJ, Kessler L, Burrows F, Elkabets M. Anti-PD1 prolongs the response of PI3K and farnesyl transferase inhibition in HRAS- and PIK3CA-mutant head and neck cancers. Neoplasia 2025; 63:101157. [PMID: 40117718 PMCID: PMC11978339 DOI: 10.1016/j.neo.2025.101157] [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: 02/23/2025] [Accepted: 03/12/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Tipifarnib, a farnesyl transferase inhibitor, has shown promising response in the treatment of HRAS-mutant HNSCC in the clinic, and in combination with a PI3K inhibitor in PIK3CA-mutant mouse models; however, the involvement of antitumor immunity in the efficacy of tipifarnib has not yet been investigated. This study aimed to evaluate the involvement of antitumor immunity in the efficacy of tipifarnib in HRAS- or PIK3CA-mutant HPV-positive and HPV-negative head and neck cancer murine models. METHODS To investigate the role of antitumor immunity, we compared the efficacy of tipifarnib in immune-intact C57BL/6 mice and immunodeficient NSG mice. Histopathological analyses were conducted to evaluate PD-L1 expression and the activation of key signaling pathways. Additionally, the synergistic potential of tipifarnib with the PI3Kα inhibitor alpelisib (BYL719) was assessed in vitro and in vivo. Immunohistochemical analysis was performed to examine the infiltration of CD8+T cells, and anti-PD1 treatment was tested to evaluate its potential to prolong progression-free survival. RESULTS In the HPV-positive HRAS-mutant HNSCC model, the antitumor efficacy of tipifarnib was primarily dependent on CD8+T cell activity, whereas in HPV-negative cancers, the contribution of antitumor immunity was less pronounced. Tipifarnib treatment upregulated PD-L1 expression, potentially inhibiting T cell antitumor activity and inducing hyperactivation of the AKT pathway, which mitigated MAPK inhibition and promoted cell proliferation. Blocking the PI3K pathway with alpelisib demonstrated synergistic antitumor effects in all models. The combination of tipifarnib and alpelisib exhibited greater efficacy in immune-intact mice than in immunodeficient mice, and was accompanied by increased CD8+T cell infiltration. Adding anti-PD1 treatment to the tipifarnib/alpelisib combination further prolonged progression-free survival in tumor-bearing mice. CONCLUSION These findings underscore the critical role of antitumor immunity, particularly CD8+T cell activity, in the efficacy of tipifarnib alone and in combination with alpelisib. The triple combination of tipifarnib, alpelisib, and anti-PD1 treatment showed superior antitumor activity and extended survival in preclinical models, suggesting its potential as a therapeutic strategy for HNSCC patients with HRAS- and PIK3CA-mutation.
Collapse
Affiliation(s)
- Dinesh Babu Manikandan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sankar Jagadeeshan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sooraj Mathukkada
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Raghda Abu Shareb
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Manu Prasad
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Liju Vijaya Steltar Belsamma
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Divyasree Marripati
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Noga Erez
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Monica Wainer
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Amit Geva
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Danielle Raviv
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Irit Allon
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Institute of Pathology, Barzilai University Medical Center, Ashqelon, Israel
| | - Luc Gt Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Gloria H Su
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology, Columbia University Medical Center, New York, NY, USA; Department of Otolaryngology/Head and Neck Surgery, Columbia University Medical Center, New York, NY, USA
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, and University of Chinese Academy of Sciences, Beijing, China
| | - Ari J Rosenberg
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | | | | | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
2
|
Wei C, Liu M, Zhang W. Programmed cell death protein 1 in cancer cells. Cell Commun Signal 2025; 23:185. [PMID: 40241148 PMCID: PMC12001728 DOI: 10.1186/s12964-025-02155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Programmed cell death protein 1 (PD-1) is frequently detected in certain subsets of tumor cells, and our understanding of PD-1 signaling consequences has expanded to include control of tumor growth, stemness and drug resistance. Nonetheless, tumor cell-intrinsic PD-1 has been comparatively underexplored in relation to PD-1 expressed on the surface of immune cells as an immune checkpoint, despite the imperative need to comprehensively elucidate the underlying mechanisms of action for achieving optimal responses in tumor immunotherapy. Here, we review the roles of the regulation and function of tumor-intrinsic PD-1 from its expression to degradation processes. Our primary focus is on unraveling its enigmatic influence on tumorigenesis and progression as proposed by recent findings, while navigating the labyrinthine network of regulatory mechanisms governing its expression and intricate functional interplay. We also discuss how the elucidation of the mechanistic underpinnings of tumor-intrinsic PD-1 expression holds the potential to explain the divergent therapeutic outcomes observed with anti-PD-1-based combination therapies, thereby furnishing indispensable insights crucial for synergistic anti-tumor strategies.
Collapse
Affiliation(s)
- Chunlian Wei
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, Shandong, PR China
- Shandong Engineering Researh Center for Smart Materials and Regenerative Medicine, Shandong Second Medical University, Weifang, 261053, Shandong, PR China
| | - Meijun Liu
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, PR China
- Shandong Engineering Researh Center for Smart Materials and Regenerative Medicine, Shandong Second Medical University, Weifang, 261053, Shandong, PR China
| | - Weifen Zhang
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, PR China.
- Shandong Engineering Researh Center for Smart Materials and Regenerative Medicine, Shandong Second Medical University, Weifang, 261053, Shandong, PR China.
| |
Collapse
|
3
|
Chaudhary PK, Kim S, Kim S. Effects and molecular mechanisms of farnesyltransferase inhibitor tipifarnib on platelet activation. J Pharmacol Exp Ther 2025; 392:103530. [PMID: 40139073 DOI: 10.1016/j.jpet.2025.103530] [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: 09/17/2024] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Tipifarnib, a farnesyltransferase inhibitor, substantially protects against cardiovascular diseases and is currently undergoing clinical trials to treat various cancers. Platelets have a well-recognized role in the progression of cancer-associated cardiovascular diseases. Nevertheless, the effect of tipifarnib on platelet function has not been studied thus far. Thus, we investigated the effect of tipifarnib and its molecular basis on the regulation of platelet activation. 2-Methylthioadenosine diphosphate (2-MeSADP)-induced secondary waves of aggregation and dense granule secretion in murine-washed platelets were completely inhibited by tipifarnib. Since 2-MeSADP-induced secretion and the resultant secondary wave of aggregation are mediated by the positive feedback effect of thromboxane A2 (TxA2) generation, it suggests the important role of tipifarnib on TxA2 generation in platelets. Consistently, tipifarnib did not affect the 2-MeSADP-induced platelet aggregation in aspirinated platelets where the contribution of TxA2 generation was blocked. In addition, platelet aggregation and secretion induced by low concentrations of AYPGKF and thrombin, which are affected by the positive feedback effect of TxA2 generation, were partially inhibited by tipifarnib. Importantly, the ELISA assay showed that 2-MeSADP- and AYPGKF-induced TxA2 generation was significantly inhibited in the presence of tipifarnib, confirming the role of tipifarnib on TxA2 generation. Finally, tipifarnib significantly inhibited 2-MeSADP-induced protein kinase B and extracellular signal-regulated kinases phosphorylation only in nonaspirinated platelets but not in aspirinated platelets, indicating the contribution of TxA2 generation. Tipifarnib plays a role in platelet function by regulating TxA2 generation, thereby indicating the possibility of using tipifarnib as a single key to treat various patients with cancer with thromboembolic complications in the future. SIGNIFICANCE STATEMENT: Farnesyltransferase inhibitor tipifarnib regulates platelet activity by inhibiting thromboxane A2 generation through the modulation of protein kinase B and extracellular signal-regulated kinase phosphorylation. Given the dual role of platelets in both thrombosis and cancer progression, tipifarnib's ability to modulate these pathways highlights its potential as a therapeutic agent in preventing thromboembolic complications in patients with cancer.
Collapse
Affiliation(s)
- Preeti K Chaudhary
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Sanggu Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.
| | - Soochong Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
4
|
Pan Y, Berkovska O, Marathe S, Mermelekas G, Gudoityte G, Wolide AD, Arslan T, Seashore-Ludlow B, Lehtiö J, Orre LM. Functional-proteomics-based investigation of the cellular response to farnesyltransferase inhibition in lung cancer. iScience 2025; 28:111864. [PMID: 39995872 PMCID: PMC11848503 DOI: 10.1016/j.isci.2025.111864] [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: 05/08/2024] [Revised: 10/16/2024] [Accepted: 01/17/2025] [Indexed: 02/26/2025] Open
Abstract
Farnesylation is a lipid post-translational modification of proteins crucial for protein membrane anchoring and cellular signaling. Farnesyltransferase inhibitors (FTIs), such as tipifarnib, are being tested in cancer therapy. However, the full impact of FTIs on farnesylation substrates remains poorly understood, thus limiting their use in precision medicine. In this study, we performed a global proteomics analysis to investigate farnesylation and the effects of tipifarnib in lung cancer cell lines. Using metabolic labeling and mass spectrometry, we identified farnesylated proteins and mapped their subcellular localization. We also analyzed tipifarnib-dependent protein relocalization and proteome-wide changes. Key findings include the potential therapeutic value of FTIs for NRAS-mutated melanoma and GNAQ/GNA11-mutated uveal melanoma by inhibiting INPP5A farnesylation. Additionally, we identified a synergistic drug combination involving tipifarnib and a ferroptosis inducer and discovered PTP4A1 as a regulator of interferon signaling. Our data, covering 15,080 proteins, offer valuable insights for future studies of farnesylation and FTIs.
Collapse
Affiliation(s)
- Yanbo Pan
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| | - Olena Berkovska
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| | - Soumitra Marathe
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| | - Georgios Mermelekas
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| | - Greta Gudoityte
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| | - Amare D. Wolide
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| | - Taner Arslan
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| | - Brinton Seashore-Ludlow
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| | - Janne Lehtiö
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| | - Lukas M. Orre
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17165 Solna, Sweden
| |
Collapse
|
5
|
Suleiman R, McGarrah P, Baral B, Owen D, Vera Aguilera J, Halfdanarson TR, Price KA, Fuentes Bayne HE. Alpelisib and Immunotherapy: A Promising Combination for Recurrent and Metastatic Squamous Cell Carcinoma of the Head and Neck. Cancer Rep (Hoboken) 2024; 7:e70023. [PMID: 39376013 PMCID: PMC11458888 DOI: 10.1002/cnr2.70023] [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: 11/17/2023] [Revised: 07/22/2024] [Accepted: 09/10/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Recurrent squamous cell carcinoma (SCC) of the head and neck (SCCHN) remains a formidable clinical challenge despite available treatments. The phosphatidylinositol 3-kinase (PI3K) pathway has been identified as a potential therapeutic target, and alpelisib, a selective PI3Kα inhibitor, has demonstrated efficacy in certain malignancies. Combining this targeted therapy with immunotherapy has been suggested in previous studies as a promising strategy to bolster the immune response against cancer. CASES A 69-year-old woman with locoregional recurrence of PIK3CA-mutated SCC of the left maxilla and cervical nodal metastases. Several chemotherapeutic regimens, including cisplatin, docetaxel, 5FU, chemoradiotherapy, and mono-immunotherapy, resulted in disease progression. Alpelisib combined with pembrolizumab led to a sustained response for 9 months. A 58-year-old man with recurrent metastatic PIK3CA-mutated SCC of the oropharynx, involving the left lung, hilar, and mediastinal lymph nodes. Despite prior palliative radiation and platinum-based chemotherapy with pembrolizumab and cetuximab, treatment with alpelisib and nivolumab resulted in a partial response. Severe hyperglycemia and rash led to treatment discontinuation. CONCLUSION Our findings highlight the potential of this innovative therapeutic combination, suggesting a need for further investigations in this setting.
Collapse
Affiliation(s)
- Riham Suleiman
- Division of Medical OncologyMayo ClinicRochesterMinnesotaUSA
| | | | - Binav Baral
- Division of Medical OncologyMayo ClinicRochesterMinnesotaUSA
| | - Dawn Owen
- Division of Radiation OncologyMayo ClinicRochesterMinnesotaUSA
| | | | | | | | | |
Collapse
|
6
|
Liu S, Han B, Wang R, Fang J. Elucidating the role of FOS in modulating the immune microenvironment through fibroblast and myeloid cell regulation in locoregional recurrent HNSCC. ENVIRONMENTAL TOXICOLOGY 2024; 39:4531-4546. [PMID: 38567514 DOI: 10.1002/tox.24262] [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: 02/05/2024] [Revised: 03/10/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) presents a significant clinical challenge, particularly due to its high propensity for locoregional recurrence. Current research underscores the need to unravel the complex interactions within the tumor microenvironment. This study addresses the critical gap in understanding how FOS modulates the immune landscape in HNSCC, with a focus on its influence on fibroblast and myeloid cell dynamics. METHODS Employing a comprehensive approach, we analyzed tissue samples from HNSCC patients and adjacent non-cancerous tissues using bulk RNA sequencing complemented by in-depth bioinformatics analyses, including gene ontology (GO), Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis, and immune infiltration assessment. A pivotal aspect of our research involved dissecting single-cell RNA-seq data from GSE234933 to elucidate the cell-type-specific expression of FOS. RESULTS We found that FOS expression varies significantly in different cell populations in the HNSCC tumor microenvironment, especially in fibroblasts and myeloid cells. This expression difference may reflect the different roles of these cells in tumor progression and their impact on the tumor microenvironment. CONCLUSION Our results uncover a significant correlation between FOS expression and key immune and hypoxia-related pathways, suggesting its integral role in the tumor microenvironment. These findings not only enhance our understanding of HNSCC pathogenesis but also highlight FOS as a potential therapeutic target. This study marks a significant step towards addressing the urgent need for targeted interventions in HNSCC, particularly in the context of locoregional recurrence.
Collapse
Affiliation(s)
- Shaokun Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Boxuan Han
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ru Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jugao Fang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Healy FM, Turner AL, Marensi V, MacEwan DJ. Mediating kinase activity in Ras-mutant cancer: potential for an individualised approach? Front Pharmacol 2024; 15:1441938. [PMID: 39372214 PMCID: PMC11450236 DOI: 10.3389/fphar.2024.1441938] [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: 05/31/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
It is widely acknowledged that there is a considerable number of oncogenic mutations within the Ras superfamily of small GTPases which are the driving force behind a multitude of cancers. Ras proteins mediate a plethora of kinase pathways, including the MAPK, PI3K, and Ral pathways. Since Ras was considered undruggable until recently, pharmacological targeting of pathways downstream of Ras has been attempted to varying success, though drug resistance has often proven an issue. Nuances between kinase pathway activation in the presence of various Ras mutants are thought to contribute to the resistance, however, the reasoning behind activation of different pathways in different Ras mutational contexts is yet to be fully elucidated. Indeed, such disparities often depend on cancer type and disease progression. However, we are in a revolutionary age of Ras mutant targeted therapy, with direct-targeting KRAS-G12C inhibitors revolutionising the field and achieving FDA-approval in recent years. However, these are only beneficial in a subset of patients. Approximately 90% of Ras-mutant cancers are not KRAS-G12C mutant, and therefore raises the question as to whether other distinct amino acid substitutions within Ras may one day be targetable in a similar manner, and indeed whether better understanding of the downstream pathways these various mutants activate could further improve therapy. Here, we discuss the favouring of kinase pathways across an array of Ras-mutant oncogenic contexts and assess recent advances in pharmacological targeting of various Ras mutants. Ultimately, we will examine the utility of individualised pharmacological approaches to Ras-mediated cancer.
Collapse
Affiliation(s)
- Fiona M. Healy
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Amy L. Turner
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Vanessa Marensi
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Chester Medical School, University of Chester, Chester, United Kingdom
| | - David J. MacEwan
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
8
|
Liu Y, Zhang N, Wen Y, Wen J. Head and neck cancer: pathogenesis and targeted therapy. MedComm (Beijing) 2024; 5:e702. [PMID: 39170944 PMCID: PMC11338281 DOI: 10.1002/mco2.702] [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: 01/30/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Head and neck cancer (HNC) is a highly aggressive type of tumor characterized by delayed diagnosis, recurrence, metastasis, relapse, and drug resistance. The occurrence of HNC were associated with smoking, alcohol abuse (or both), human papillomavirus infection, and complex genetic and epigenetic predisposition. Currently, surgery and radiotherapy are the standard treatments for most patients with early-stage HNC. For recurrent or metastatic (R/M) HNC, the first-line treatment is platinum-based chemotherapy combined with the antiepidermal growth factor receptor drug cetuximab, when resurgery and radiation therapy are not an option. However, curing HNC remains challenging, especially in cases with metastasis. In this review, we summarize the pathogenesis of HNC, including genetic and epigenetic changes, abnormal signaling pathways, and immune regulation mechanisms, along with all potential therapeutic strategies such as molecular targeted therapy, immunotherapy, gene therapy, epigenetic modifications, and combination therapies. Recent preclinical and clinical studies that may offer therapeutic strategies for future research on HNC are also discussed. Additionally, new targets and treatment methods, including antibody-drug conjugates, photodynamic therapy, radionuclide therapy, and mRNA vaccines, have shown promising results in clinical trials, offering new prospects for the treatment of HNC.
Collapse
Affiliation(s)
- Yan Liu
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengduChina
- National Facility for Translational Medicine (Sichuan)West China Hospital of Sichuan UniversityChengduChina
| | - Nannan Zhang
- National Center for Birth Defect MonitoringKey Laboratory of Birth Defects and Related Diseases of Women and ChildrenMinistry of EducationWest China Second University HospitalSichuan UniversityChengduChina
| | - Yi Wen
- State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Jiaolin Wen
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengduChina
| |
Collapse
|
9
|
Okudaira N, Burt H, Mitra A. Tipifarnib physiologically-based pharmacokinetic modeling to assess drug-drug interaction, organ impairment, and biopharmaceutics in healthy subjects and cancer patients. CPT Pharmacometrics Syst Pharmacol 2024; 13:1366-1379. [PMID: 38807307 PMCID: PMC11330181 DOI: 10.1002/psp4.13165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 05/30/2024] Open
Abstract
A physiologically-based pharmacokinetic (PBPK) model for tipifarnib, which included mechanistic absorption, was built and verified by integrating in vitro data and several clinical data in healthy subjects and cancer patients. The final PBPK model was able to recover the clinically observed single and multiple-dose plasma concentrations of tipifarnib in healthy subjects and cancer patients under several dosing conditions, such as co-administration with a strong CYP3A4 inhibitor and inducer, an acid-reducing agent (proton pump inhibitor and H2 receptor antagonist), and with a high-fat meal. In addition, the model was able to accurately predict the effect of mild or moderate hepatic impairment on tipifarnib exposure. The appropriately verified model was applied to prospectively simulate the liability of tipifarnib as a victim of CYP3A4 enzyme-based drug-drug interactions (DDIs) with a moderate inhibitor and inducer as well as tipifarnib as a perpetrator of DDIs with sensitive substrates of CYP3A4, CYP2B6, CYP2D6, CYP2C9, and CYP2C19 in healthy subjects and cancer patients. The effect of a high-fat meal, acid-reducing agent, and formulation change at the therapeutic dose was simulated. Finally, the model was used to predict the effect of mild, moderate, or severe hepatic, and renal impairment on tipifarnib PK. This multipronged approach of combining the available clinical data with PBPK modeling-guided dosing recommendations for tipifarnib under several conditions. This example showcases the totality of the data approach to gain a more thorough understanding of clinical pharmacology and biopharmaceutic properties of oncology drugs in development.
Collapse
Affiliation(s)
| | - Howard Burt
- Certara, UK Ltd. (Simcyp Division)SheffieldUK
| | - Amitava Mitra
- Clinical PharmacologyKura Oncology, Inc.BostonMassachusettsUSA
| |
Collapse
|
10
|
Mallaredy V, Roy R, Cheng Z, Gurrala CT, Benedict C, Truongcao M, Joladarashi D, Magadum A, Ibetti J, Cimini M, Gonzalez C, Garikipati VNS, Koch WJ, Kishore R. Tipifarnib Reduces Extracellular Vesicles and Protects From Heart Failure. Circ Res 2024; 135:280-297. [PMID: 38847080 PMCID: PMC11223950 DOI: 10.1161/circresaha.123.324110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/28/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Heart failure (HF) is one of the leading causes of mortality worldwide. Extracellular vesicles, including small extracellular vesicles or exosomes, and their molecular cargo are known to modulate cell-to-cell communication during multiple cardiac diseases. However, the role of systemic extracellular vesicle biogenesis inhibition in HF models is not well documented and remains unclear. METHODS We investigated the role of circulating exosomes during cardiac dysfunction and remodeling in a mouse transverse aortic constriction (TAC) model of HF. Importantly, we investigate the efficacy of tipifarnib, a recently identified exosome biogenesis inhibitor that targets the critical proteins (Rab27a [Ras associated binding protein 27a], nSMase2 [neutral sphingomyelinase 2], and Alix [ALG-2-interacting protein X]) involved in exosome biogenesis for this mouse model of HF. In this study, 10-week-old male mice underwent TAC surgery were randomly assigned to groups with and without tipifarnib treatment (10 mg/kg 3 times/wk) and monitored for 8 weeks, and a comprehensive assessment was conducted through performed echocardiographic, histological, and biochemical studies. RESULTS TAC significantly elevated circulating plasma exosomes and markedly increased cardiac left ventricular dysfunction, cardiac hypertrophy, and fibrosis. Furthermore, injection of plasma exosomes from TAC mice induced left ventricular dysfunction and cardiomyocyte hypertrophy in uninjured mice without TAC. On the contrary, treatment of tipifarnib in TAC mice reduced circulating exosomes to baseline and remarkably improved left ventricular functions, hypertrophy, and fibrosis. Tipifarnib treatment also drastically altered the miRNA profile of circulating post-TAC exosomes, including miR 331-5p, which was highly downregulated both in TAC circulating exosomes and in TAC cardiac tissue. Mechanistically, miR 331-5p is crucial for inhibiting the fibroblast-to-myofibroblast transition by targeting HOXC8, a critical regulator of fibrosis. Tipifarnib treatment in TAC mice upregulated the expression of miR 331-5p that acts as a potent repressor for one of the fibrotic mechanisms mediated by HOXC8. CONCLUSIONS Our study underscores the pathological role of exosomes in HF and fibrosis in response to pressure overload. Tipifarnib-mediated inhibition of exosome biogenesis and cargo sorting may serve as a viable strategy to prevent progressive cardiac remodeling in HF.
Collapse
Affiliation(s)
- Vandana Mallaredy
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - Rajika Roy
- Department of Surgery, Division of Cardiovascular and Thoracic Surgery, Duke University School of Medicine, Durham, NC 27710
| | - Zhongjian Cheng
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - Charan Thej Gurrala
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - Cindy Benedict
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - May Truongcao
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - Darukeshwara Joladarashi
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - Ajit Magadum
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - Jessica Ibetti
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - Maria Cimini
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - Carolina Gonzalez
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
| | - Venkata Naga Srikanth Garikipati
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140
| | - Walter J. Koch
- Department of Surgery, Division of Cardiovascular and Thoracic Surgery, Duke University School of Medicine, Durham, NC 27710
| | - Raj Kishore
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140
| |
Collapse
|
11
|
Yue Z, Cheng K. "Tip" the Scale of Cardiac Repair via Reducing Pathological Extracellular Vesicles. Circ Res 2024; 135:298-300. [PMID: 38963869 PMCID: PMC11285627 DOI: 10.1161/circresaha.124.324955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Affiliation(s)
- Zhang Yue
- Department of Biomedical Engineering, Columbia University,
New York, NY, USA
- Department of Cardiovascular Surgery, Union Hospital,
Tongji Medical College, Huazhong University of Science and Technology, Wuhan,
China
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University,
New York, NY, USA
| |
Collapse
|
12
|
Ibanez KR, Huang TT, Lee JM. Combination Therapy Approach to Overcome the Resistance to PI3K Pathway Inhibitors in Gynecological Cancers. Cells 2024; 13:1064. [PMID: 38920692 PMCID: PMC11201409 DOI: 10.3390/cells13121064] [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: 05/29/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
The PI3K signaling pathway plays an essential role in cancer cell proliferation and survival. PI3K pathway inhibitors are now FDA-approved as a single agent treatment or in combination for solid tumors such as renal cell carcinoma or breast cancer. However, despite the high prevalence of PI3K pathway alterations in gynecological cancers and promising preclinical activity in endometrial and ovarian cancer models, PI3K pathway inhibitors showed limited clinical activity in gynecological cancers. In this review, we provide an overview on resistance mechanisms against PI3K pathway inhibitors that limit their use in gynecological malignancies, including genetic alterations that reactivate the PI3K pathway such as PIK3CA mutations and PTEN loss, compensatory signaling pathway activation, and feedback loops causing the reactivation of the PI3K signaling pathway. We also discuss the successes and limitations of recent clinical trials aiming to address such resistance mechanisms through combination therapies.
Collapse
|
13
|
Ding M, Gao J, Wang J, Li Z, Gong X, Cui Z, Li C, Xue H, Li D, Wang Y. Colorectal cancer subtyping and immune landscape analysis based on natural killer cell-related genes. Arab J Gastroenterol 2024; 25:150-159. [PMID: 38719664 DOI: 10.1016/j.ajg.2024.03.005] [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: 07/13/2023] [Revised: 01/29/2024] [Accepted: 03/20/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND AND STUDY AIMS The prognosis of colorectal cancer (CRC) is related to natural killer (NK) cells, but the molecular subtype features of CRC based on NK cells are still unknown. This study aimed to identify NK cell-related molecular subtypes of CRC and analyze the survival status and immune landscape of patients with different subtypes. PATIENTS/MATERIAL AND METHODS mRNA expression data, single nucleotide variant (SNV) data, and clinical information of CRC patients were obtained from The Cancer Genome Atlas. Differentially expressed genes (DEGs) were obtained through differential analysis, and the intersection was taken with NK cell-associated genes to obtain 103 NK cell-associated CRC DEGs (NCDEGs). Based on NCDEGs, CRC samples were divided into three clusters through unsupervised clustering analysis. Survival analysis, immune analysis, Gene Set Enrichment Analysis (GSEA), and tumor mutation burden (TMB) analysis were performed. Finally, NCDEG-related small-molecule drugs were screened using the CMap database. RESULTS Survival analysis revealed that cluster2 had a lower survival rate than cluster1 and cluster3 (p < 0.05). Immune infiltration analysis found that the immune infiltration levels and immune checkpoint expression levels of cluster1_3 were substantially higher than those of cluster2, and the tumor purity was the opposite (p < 0.05). GSEA presented that cluster1_3 was significantly enriched in the chemokine signaling pathway, ECM receptor interaction, and antigen processing and presentation pathways (p < 0.05). The TMB of cluster1_3 was significantly higher than that of cluster2 (p < 0.05). Genes with the highest mutation rate in CRC were APC, TP53, TTN, and KRAS. Drug prediction results showed that small-molecule drugs that reverse the upregulation of NCDEGs, deoxycholic acid, dipivefrine, phenformin, and other drugs may improve the prognosis of CRC. CONCLUSION NK cell-associated CRC subtypes can be used to evaluate the tumor characteristics of CRC patients and provide an important reference for CRC patients.
Collapse
Affiliation(s)
- Mei Ding
- Surgical Research Division, Tangshan Vocational & Technical College, Tangshan, Hebei 063000, China; Department of Laparoscopy and Colorectal Surgery, Tangshan Central Hospital, Tangshan, Hebei 063000, China
| | - Jianchao Gao
- Department of Laparoscopy and Colorectal Surgery, Tangshan Central Hospital, Tangshan, Hebei 063000, China
| | - Jinyan Wang
- Department of Laparoscopy and Colorectal Surgery, Tangshan Central Hospital, Tangshan, Hebei 063000, China
| | - Zongfu Li
- Surgical Research Division, Tangshan Vocational & Technical College, Tangshan, Hebei 063000, China
| | - Xiangliang Gong
- Department of Laparoscopy and Colorectal Surgery, Tangshan Central Hospital, Tangshan, Hebei 063000, China
| | - Zhiyu Cui
- Department of Laparoscopy and Colorectal Surgery, Tangshan Central Hospital, Tangshan, Hebei 063000, China
| | - Changjun Li
- Department of Laparoscopy and Colorectal Surgery, Tangshan Central Hospital, Tangshan, Hebei 063000, China
| | - Hongjun Xue
- Department of Laparoscopy and Colorectal Surgery, Tangshan Central Hospital, Tangshan, Hebei 063000, China
| | - Dandan Li
- Department of Pathology, Tangshan Central Hospital, Tangshan, Hebei 063000, China
| | - Yigang Wang
- Department of Laparoscopy and Colorectal Surgery, Tangshan Central Hospital, Tangshan, Hebei 063000, China.
| |
Collapse
|
14
|
Tate EW, Soday L, de la Lastra AL, Wang M, Lin H. Protein lipidation in cancer: mechanisms, dysregulation and emerging drug targets. Nat Rev Cancer 2024; 24:240-260. [PMID: 38424304 DOI: 10.1038/s41568-024-00666-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2024] [Indexed: 03/02/2024]
Abstract
Protein lipidation describes a diverse class of post-translational modifications (PTMs) that is regulated by over 40 enzymes, targeting more than 1,000 substrates at over 3,000 sites. Lipidated proteins include more than 150 oncoproteins, including mediators of cancer initiation, progression and immunity, receptor kinases, transcription factors, G protein-coupled receptors and extracellular signalling proteins. Lipidation regulates the physical interactions of its protein substrates with cell membranes, regulating protein signalling and trafficking, and has a key role in metabolism and immunity. Targeting protein lipidation, therefore, offers a unique approach to modulate otherwise undruggable oncoproteins; however, the full spectrum of opportunities to target the dysregulation of these PTMs in cancer remains to be explored. This is attributable in part to the technological challenges of identifying the targets and the roles of protein lipidation. The early stage of drug discovery for many enzymes in the pathway contrasts with efforts for drugging similarly common PTMs such as phosphorylation and acetylation, which are routinely studied and targeted in relevant cancer contexts. Here, we review recent advances in identifying targetable protein lipidation pathways in cancer, the current state-of-the-art in drug discovery, and the status of ongoing clinical trials, which have the potential to deliver novel oncology therapeutics targeting protein lipidation.
Collapse
Affiliation(s)
- Edward W Tate
- Department of Chemistry, Imperial College London, London, UK.
- Francis Crick Institute, London, UK.
| | - Lior Soday
- Department of Chemistry, Imperial College London, London, UK
| | | | - Mei Wang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
| | - Hening Lin
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY, USA
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|
15
|
Jagadeeshan S, Suryamohan K, Shin N, Mathukkada S, Boyko A, Melikhova D, Tsareva A, Yunusova L, Pravdivtseva E, Stupichev D, Shaposhnikov K, Peterson A, Bednyagin L, Shugaev-Mendosa E, Kessler L, Burrows F, Ho AL, Agrawal N, Pearson AT, Izumchenko E, Cole G, Elkabets M, Rosenberg AJ. Evolutionary dynamics of tipifarnib in HRAS mutated head and neck squamous cell carcinoma. Oral Oncol 2024; 149:106688. [PMID: 38219706 PMCID: PMC11309563 DOI: 10.1016/j.oraloncology.2024.106688] [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: 09/24/2023] [Revised: 12/05/2023] [Accepted: 01/08/2024] [Indexed: 01/16/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly prevalent malignancy worldwide, with a significant proportion of patients developing recurrent and/or metastatic (R/M) disease. Despite recent advances in therapy, the prognosis for patients with advanced HNSCC remains poor. Here, we present the case of a patient with recurrent metastatic HNSCC harboring an HRAS G12S mutation who achieved a durable response to treatment with tipifarnib, a selective inhibitor of farnesyltransferase. The patient was a 48-year-old woman who had previously received multiple lines of therapy with no significant clinical response. However, treatment with tipifarnib resulted in a durable partial response that lasted 8 months. Serial genomic and transcriptomic analyses demonstrated upregulation of YAP1 and AXL in metastatic lesions compared with the primary tumor, the evolution of the tumor microenvironment from an immune-enriched to a fibrotic subtype with increased angiogenesis, and activation of the PI3K/AKT/mTOR pathway in tipifarnib treatment. Lastly, in HRAS-mutated PDXs and in the syngeneic HRAS model, we demonstrated that tipifarnib efficacy is limited by activation of the AKT pathway, and dual treatment with tipifarnib and the PI3K inhibitor, BYL719, resulted in enhanced anti-tumor efficacy. Our case study highlights the potential of targeting HRAS mutations with tipifarnib in R/M HNSCC and identifies potential mechanisms of acquired resistance to tipifarnib, along with immuno-, chemo-, and radiation therapy. Preclinical results provide a firm foundation for further investigation of drug combinations of HRAS-and PI3K -targeting therapeutics in R/M HRAS-driven HNSCC.
Collapse
Affiliation(s)
- Sankar Jagadeeshan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Nara Shin
- BostonGene Corporation, Waltham, MA, USA
| | - Sooraj Mathukkada
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | - Alan L Ho
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York City, NY, USA
| | - Nishant Agrawal
- Department of Surgery, Section of Otolaryngology-Head and Neck Surgery, University of Chicago, IL, USA
| | - Alexander T Pearson
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Grayson Cole
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Ari J Rosenberg
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
16
|
Wang Y, Fleishman JS, Li T, Li Y, Ren Z, Chen J, Ding M. Pharmacological therapy of metabolic dysfunction-associated steatotic liver disease-driven hepatocellular carcinoma. Front Pharmacol 2024; 14:1336216. [PMID: 38313077 PMCID: PMC10834746 DOI: 10.3389/fphar.2023.1336216] [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: 11/10/2023] [Accepted: 12/31/2023] [Indexed: 02/06/2024] Open
Abstract
In light of a global rise in the number of patients with type 2 diabetes mellitus (T2DM) and obesity, non-alcoholic fatty liver disease (NAFLD), now known as metabolic dysfunction-associated fatty liver disease (MAFLD) or metabolic dysfunction-associated steatotic liver disease (MASLD), has become the leading cause of hepatocellular carcinoma (HCC), with the annual occurrence of MASLD-driven HCC expected to increase by 45%-130% by 2030. Although MASLD has become a serious major public health threat globally, the exact molecular mechanisms mediating MASLD-driven HCC remain an open problem, necessitating future investigation. Meanwhile, emerging studies are focusing on the utility of bioactive compounds to halt the progression of MASLD to MASLD-driven HCC. In this review, we first briefly review the recent progress of the possible mechanisms of pathogenesis and progression for MASLD-driven HCC. We then discuss the application of bioactive compounds to mitigate MASLD-driven HCC through different modulatory mechanisms encompassing anti-inflammatory, lipid metabolic, and gut microbial pathways, providing valuable information for future treatment and prevention of MASLD-driven HCC. Nonetheless, clinical research exploring the effectiveness of herbal medicines in the treatment of MASLD-driven HCC is still warranted.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Joshua S. Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Tongda Li
- Department of Traditional Chinese Medicine, Beijing Geriatric Hospital, Beijing, China
| | - Yulin Li
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Zhao Ren
- Department of Pharmacy, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| | - Mingchao Ding
- Department of Peripheral Vascular Intervention, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, China
| |
Collapse
|
17
|
Jagadeeshan S, Novoplansky OZ, Cohen O, Kurth I, Hess J, Rosenberg AJ, Grandis JR, Elkabets M. New insights into RAS in head and neck cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188963. [PMID: 37619805 PMCID: PMC11815531 DOI: 10.1016/j.bbcan.2023.188963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
RAS genes are known to be dysregulated in cancer for several decades, and substantial effort has been dedicated to develop agents that reduce RAS expression or block RAS activation. The recent introduction of RAS inhibitors for cancer patients highlights the importance of comprehending RAS alterations in head and neck cancer (HNC). In this regard, we examine the published findings on RAS alterations and pathway activations in HNC, and summarize their role in HNC initiation, progression, and metastasis. Specifically, we focus on the intrinsic role of mutated-RAS on tumor cell signaling and its extrinsic role in determining tumor-microenvironment (TME) heterogeneity, including promoting angiogenesis and enhancing immune escape. Lastly, we summarize the intrinsic and extrinsic role of RAS alterations on therapy resistance to outline the potential of targeting RAS using a single agent or in combination with other therapeutic agents for HNC patients with RAS-activated tumors.
Collapse
Affiliation(s)
- Sankar Jagadeeshan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel.
| | - Ofra Z Novoplansky
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel.
| | - Oded Cohen
- Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel; Department of Otolaryngology- Head and Neck Surgery and Oncology, Soroka Medical Center, Beersheva, Israel.
| | - Ina Kurth
- Division of Radiooncology-Radiobiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany; Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Ari J Rosenberg
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA.
| | - Jennifer R Grandis
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, CA, USA.
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel; Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, Beer-Sheva 8410501, Israel.
| |
Collapse
|
18
|
Wang J, Al-Majid D, Brenner JC, Smith JD. Mutant HRas Signaling and Rationale for Use of Farnesyltransferase Inhibitors in Head and Neck Squamous Cell Carcinoma. Target Oncol 2023; 18:643-655. [PMID: 37665491 DOI: 10.1007/s11523-023-00993-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are often associated with poor outcomes, due at least in part to the limited number of treatment options available for those patients who develop recurrent and/or metastatic disease (R/M HNSCC). Even with the recent validation and approval of immunotherapies in the first-line setting for these patients, the need for the development of new and alternative precision medicine strategies with survival benefit is clear. Oncogenic alterations in the HRAS (Harvey rat sarcoma virus) proto-oncogene are seen in approximately 4-8% of R/M HNSCC tumors. Recently, several preclinical and clinical advancements have been made in the implementation of small-molecule inhibitors that block post-translational farnesylation of HRas, thereby abrogating its downstream oncogenic activity. In this review, we focus on the biology of wild-type and mutant HRas signaling in HNSCC, and rationale for use and outcomes of farnesyltransferase inhibitors in patients with HRAS-mutant tumors.
Collapse
Affiliation(s)
- Jiayu Wang
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dana Al-Majid
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, MSRB III 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA
| | - J Chad Brenner
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, MSRB III 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Joshua D Smith
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, MSRB III 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA
| |
Collapse
|