1
|
Fleischmann M, Kristen A, Winkelmann R, Burck I, Weigert A, Issing C, Diefenhardt M, Martin D, Brandts C, Rödel F, Balermpas P, von der Grün J, Thönissen P, Rödel C, Ghanaati S. Neoadjuvant chemoradiation for patients with resectable locally advanced oral cavity squamous cell carcinoma: Results of the prospective INVERT clinical trial. Oral Oncol 2025; 164:107252. [PMID: 40147285 DOI: 10.1016/j.oraloncology.2025.107252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE The INVERT trial is a prospective, monocentric, proof-of-principle phase 2clinical trial evaluating the safety and efficacy of neoadjuvant chemoradiation (CRT) inpatients with resectable, locally advanced oral cavity squamous cell carcinoma (LAOCSCC). METHODS Eligible patients received intensity-modulated radiotherapy (IMRT) to primarytumor and nodal volumes (60/54/50 Gy in 30 fractions) with concomitant platinumbasedchemotherapy. Surgery was performed 6-8 weeks after CRT. Primary endpointwas the pathological complete response (pCR = ypT0N0) rate. Secondary endpointsincluded R0 resection rate, surgical complications, oncological outcomes, and qualityof life. RESULTS Between 02/2012 and 09/2022, 28 UICC Stage (7th edition) IVA-IVB LAOCSCCpatients were enrolled; 92.1 % showed mandibular involvement. All patientsreceived the the full IMRT dose of 60 Gy. For all cisplatin-eligible patients, the mediancumulative dose was 200 mg/m2 BSA. In the intention-to-treat population, the pCR ratewas 46.4 % (n = 13) but did not met the primary endpoint. In total, we recorded 384adverse events (AEs) of any grade and 109 AEs ≥ grade 3 according to CTCAE (nograde 5). Surgical complications were primarily wound healing disorders and postbleeding, with no grade 4 or 5 events. At a median follow-up of 28.5 months,progression-free survival (PFS), local control (LC), freedom from distant failure (DF),and overall survival (OS) were 60.7 %, 89.3 %, 82.1 %, and 64.3 %, respectively. pCRPowered by Editorial Manager® and ProduXion Manager® from Aries Systems Corporationsignificantly correlated with improved PFS (p = 0.010) and OS (p = 0.021). CONCLUSION Neoadjuvant CRT is feasible and safe, achieving high pCR rates in LAOCSCC.pCR was associated with improved outcomes.
Collapse
Affiliation(s)
- Maximilian Fleischmann
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany.
| | - Alexander Kristen
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Ria Winkelmann
- Senckenberg Institute of Pathology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Iris Burck
- Department of Diagnostic and Interventional Radiology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Christian Issing
- Department of Otorhinolaryngology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Markus Diefenhardt
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt a. M., Goethe-University Frankfurt, Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Goethe-University Frankfurt, Frankfurt, Germany
| | - Daniel Martin
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt a. M., Goethe-University Frankfurt, Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Goethe-University Frankfurt, Frankfurt, Germany
| | - Christian Brandts
- German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt a. M., Goethe-University Frankfurt, Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Goethe-University Frankfurt, Frankfurt, Germany; Department of Medicine, Hematology/Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt a. M., Goethe-University Frankfurt, Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Goethe-University Frankfurt, Frankfurt, Germany
| | - Panagiotis Balermpas
- Department of Medicine, Hematology/Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Jens von der Grün
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Philipp Thönissen
- Department of Oral, Maxillofacial and Facial Plastic Surgery, Goethe-University Frankfurt, Frankfurt, Germany
| | - Claus Rödel
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt a. M., Goethe-University Frankfurt, Frankfurt, Germany; Frankfurt Cancer Institute (FCI), Goethe-University Frankfurt, Frankfurt, Germany
| | - Shahram Ghanaati
- Department of Oral, Maxillofacial and Facial Plastic Surgery, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
2
|
Sobti A, Skinner H, Wilke CT. Predictors of Radiation Resistance and Novel Radiation Sensitizers in Head and Neck Cancers: Advancing Radiotherapy Efficacy. Semin Radiat Oncol 2025; 35:224-242. [PMID: 40090749 DOI: 10.1016/j.semradonc.2025.02.008] [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: 10/15/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/18/2025]
Abstract
Radiation resistance in head and neck squamous cell carcinoma (HNSCC), driven by intrinsic and extrinsic factors, poses a significant challenge in radiation oncology. The key contributors are tumor hypoxia, cancer stem cells, cell cycle checkpoint activation, and DNA repair processes (homologous recombination and non-homologous end-joining). Genetic modifications such as TP53 mutations, KRAS mutations, EGFR overexpression, and abnormalities in DNA repair proteins like BRCA1/2 additionally affect radiation sensitivity. Novel radiosensitizers targeting these pathways demonstrate the potential to overcome resistance. Hypoxia-activated drugs and gold nanoparticles enhance the efficacy of radiotherapy and facilitate targeted distribution. Integrating immunotherapy, especially immune checkpoint inhibitors, with radiation therapy, enhances anti-tumor responses and reduces resistance. Epigenetic alterations, such as DNA methylation and histone acetylation, significantly influence radiation response, with the potential for sensitization through histone deacetylase inhibitors and non-coding RNA regulators. Metabolic changes linked to glucose, lipid, and glutamine metabolism influence radiosensitivity, uncovering new targets for radiosensitization. Human papillomavirus (HPV)-associated malignancies exhibit increased radiosensitivity relative to other tumors due to impaired DNA repair mechanisms and heightened immunogenicity. Furthermore, understanding the interplay between HPV oncoproteins and p53 functionality can enhance treatment strategies for HPV-related cancers. Using DNA damage response inhibitors (PARP, ATM/ATR), cell cycle checkpoint inhibitors (WEE1, CHK1/2), and hypoxia-targeted agents as radiosensitizing strategies exhibit considerable promise. Immunomodulatory approaches, including PD-1 and CTLA-4 inhibitors in conjunction with radiation, enhance anti-tumor immunity. Future directions emphasize personalized radiation therapy using genetics, sophisticated medication delivery systems, adaptive radiotherapy, and real-time monitoring. These integrated strategies seek to diminish radiation resistance and improve therapeutic efficacy in HNSCC.
Collapse
Affiliation(s)
- Aastha Sobti
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Heath Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Christopher T Wilke
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA..
| |
Collapse
|
3
|
Courtney PT, Venkat PS, Shih YCT, Chang AJ, Lee A, Steinberg ML, Raldow AC. Cost-Effectiveness of Pembrolizumab With Chemoradiotherapy for Locally Advanced Cervical Cancer. JAMA Netw Open 2025; 8:e250033. [PMID: 40036034 PMCID: PMC11880949 DOI: 10.1001/jamanetworkopen.2025.0033] [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] [Received: 07/23/2024] [Accepted: 12/19/2024] [Indexed: 03/06/2025] Open
Abstract
Importance The KEYNOTE-A18 trial demonstrated that adding concurrent and adjuvant pembrolizumab to chemoradiotherapy and brachytherapy significantly improved survival in patients with newly diagnosed, locally advanced cervical cancer. However, considering the annual global incidence of 660 000 cases of cervical cancer, including 13 820 in the US in 2024, incorporating this regimen into the standard of care could have substantial health care economic implications for both patients and the health care system. Objective To determine the cost-effectiveness of adding pembrolizumab to the first-line treatment of newly diagnosed, locally advanced cervical cancer. Design, Setting, and Participants This economic evaluation created a Markov model simulating 50-year outcomes to evaluate cost-effectiveness from the payer perspective for patients receiving either pembrolizumab or placebo in addition to chemoradiotherapy plus brachytherapy. Probabilities, including disease progression, survival, and treatment-related toxic effects, were derived from KEYNOTE-A18 clinical trial data in patients with newly diagnosed, locally advanced cervical cancer. Costs and health utilities were obtained from published literature; 1-way, 3-way, and probabilistic sensitivity analyses were used to assess model uncertainty. Data analyses were conducted from April to November 2024. Exposure Pembrolizumab. Main Outcomes and Measures Costs, measured in 2024 US dollars, and effectiveness, measured in quality-adjusted life-years (QALYs) were used to calculate an incremental cost-effectiveness ratio (ICER). A willingness-to-pay threshold of $100 000 per QALY was chosen, below which pembrolizumab would be considered cost-effective. Results KEYNOTE-A18 enrolled 1060 patients (529 in pembrolizumab group, 531 in placebo group). The median age was 50 years. Pembrolizumab increased costs by $257 000 and effectiveness by 1.40 QALYs, yielding an incremental cost-effectiveness ratio of $183 400 per QALY. The addition of pembrolizumab became cost-effective if its monthly cost was decreased from $16 990 to $9190 (a 45.6% reduction) or its maximum duration of 24 months was decreased to 10 months. The model was insensitive to assumptions about treatment-related toxic effects, progression-free survival, and overall survival. Probabilistic sensitivity analysis indicated that at a willingness-to-pay threshold of $100 000 per QALY, the addition of pembrolizumab was cost-effective 37.3% of the time. Conclusions and Relevance In this economic evaluation of adding concurrent and adjuvant pembrolizumab to first-line treatment of newly diagnosed, locally advanced cervical cancer, this regimen was not cost-effective at current prices despite data demonstrating improved survival with this regimen.
Collapse
Affiliation(s)
| | - Puja S. Venkat
- Department of Radiation Oncology, University of California, Los Angeles
| | - Ya-Chen Tina Shih
- Department of Radiation Oncology, University of California, Los Angeles
| | - Albert J. Chang
- Department of Radiation Oncology, University of California, Los Angeles
| | - Alan Lee
- Department of Radiation Oncology, University of California, Los Angeles
| | | | - Ann C. Raldow
- Department of Radiation Oncology, University of California, Los Angeles
| |
Collapse
|
4
|
Kim SG, Tasoulas J, Sheth S, Yarbrough WG, Hackman T, Amelio AL, Sullivan CB. The Role of Immunotherapy in Salivary Gland Cancer: A Systematic Review. EAR, NOSE & THROAT JOURNAL 2025:1455613251324353. [PMID: 40018983 DOI: 10.1177/01455613251324353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025] Open
Abstract
Background: Salivary gland cancer (SGC) comprises a very heterogeneous group of rare neoplasms, accounting for approximately 2% to 6% of head and neck malignancies. Surgery with or without adjuvant radiotherapy is the main treatment regimen. However, patients who are ineligible for surgery, including late-stage, recurrent, or metastatic disease, have very limited options. Chemotherapeutic schemes have failed to offer meaningful survival benefit, while the role of immune checkpoint inhibitors (ICIs) like anti-PD1, anti-PDL1, and anti-CTLA4 is largely unknown. Methods: A systematic database search of clinical trials evaluating the role of anti-PD1 and anti-CTLA4 immunotherapy in the survival of patients with SGC was conducted in EMBASE, MEDLINE, and Scopus databases. Primary outcomes were overall survival, progression-free survival, complete responses (CRs), partial responses (PRs), stable disease (SD), and objective response rates. Results: We identified 770 relevant studies. Nine clinical trials and 4 retrospective studies met the inclusion criteria and were eligible for further analysis. A total of 473 patients were studied, with an average age of 61 ± 4 years old for prospective trials and 60 ± 11 years old for retrospective studies. For studies that provided gender, the male-to-female ratio was 1.2:1 for prospective trials versus 4:1 for retrospective studies. All patients in the clinical trials had recurrent or metastatic disease. All patients received anti-PD1 ICI with either pembrolizumab or nivolumab, 7 study arms administering pembrolizumab, 10 study arms administering nivolumab. For prospective trials, 6/9 studies also reported an additional intervention. Most prevalent histology was adenoid cystic carcinoma (n = 230). Outcome of prospective trial were 1 CR, 19 PRs 145 SD, and 80% of patients reporting an adverse event (AE) of any grade. For retrospective studies, 1 patient reported CR, 3 patients reported PR, 11 patients reported SD, and 92% of patients reported an AE of any grade. Conclusion: Anti-PD1 immunotherapeutic modalities can be a safe and potentially-beneficial option for patients with advanced, recurrent, or metastatic SGC. However, the literature suffers from small cohorts, lack of randomization, and heterogeneity among different histologies of SGC. Prospective trials evaluating the role of anti-PD1 in patients with SGC, stratified by histology are warranted to determine the potential role of immunotherapy in the treatment of this disease.
Collapse
Affiliation(s)
- Sul Gi Kim
- Department of Otolaryngology-Head and Neck Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jason Tasoulas
- Department of Otolaryngology-Head and Neck Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Siddharth Sheth
- Division of Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wendell G Yarbrough
- Department of Otolaryngology-Head and Neck Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Trevor Hackman
- Department of Otolaryngology-Head and Neck Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Antonio L Amelio
- Department of Otolaryngology-Head and Neck Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center, Tampa, FL, USA
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Christopher Blake Sullivan
- Department of Otolaryngology-Head and Neck Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
5
|
Haider M, Jagal J, Ali Alghamdi M, Haider Y, Hassan HAFM, Najm MB, Jayakuma MN, Ezzat H, Greish K. Erlotinib and curcumin-loaded nanoparticles embedded in thermosensitive chitosan hydrogels for enhanced treatment of head and neck cancer. Int J Pharm 2024; 666:124825. [PMID: 39401579 DOI: 10.1016/j.ijpharm.2024.124825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/20/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) remain a major oncological challenge with significant morbidity and mortality rates. Erlotinib (Er) and Curcumin (Cm) are potential therapeutic agents for HNSCC, yet they are hindered by poor solubility and bioavailability. This study explored the optimization of poly(lactic-co-glycolic acid) nanoparticles co-loaded with Er and Cm (Er/Cm-NP), prepared via a D-optimal response surface design-guided nanoprecipitation process. The optimized formulation, optEr/Cm-NP, was then incorporated into chitosan/β-glycerophosphate hydrogels (optEr/Cm-NP-HG) to create an injectable intratumoral (IT) nanocomposite hydrogel (HG) delivery system. Physicochemical properties of the formulations, including gelation time, injectability, mechanical strength and drug release profiles were assessed alongside hemolytic activity. Compared to optEr/Cm-NP alone, the NP-loaded HG formulation exhibited a more pronounced modulation effect, enabling sustained and controlled drug release. The cytotoxicity of the developed formulations was evaluated using the FaDu HNSCC cancer cell line. Both optEr/Cm-NP and optEr/Cm-NP-HG21 displayed enhanced cytotoxicity compared to free drugs. Confocal laser microscopy and flow cytometry confirmed superior cellular uptake of Er and Cm when delivered via NPs or NP-loaded HG. Furthermore, a significant increase in apoptotic cell death upon treatment with optEr/Cm-NP was observed, highlighting its potential for HNSCC therapy. In vivo studies conducted on a xenograft HNSCC mouse model revealed the significant capacity of the intratumorally-injected optEr/Cm-NP-HG21 formulation to retard the tumor growth. Conclusively, the results presented herein report the successful development of a nanocomposite HG system incorporating NPs co-loaded with Er and Cm that could be efficiently utilized in the treatment of HNSCC.
Collapse
Affiliation(s)
- Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, 27272 Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| | - Jayalakshmi Jagal
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Maha Ali Alghamdi
- Department of Biotechnology, College of Science, Taif University, Taif 21974, Kingdom of Saudi Arabia; Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences Arabian Gulf University, Manama 329, Bahrain
| | - Youssef Haider
- College of Engineering, Boston University, Boston, MA, USA
| | - Hatem A F M Hassan
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, Canterbury ME4 4TB, UK; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Muna B Najm
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Manju N Jayakuma
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Helal Ezzat
- Research Institute of Sciences and Engineering, University of Sharjah, 27272, Sharjah, United Arab Emirates; Civil Engineering Department, Delta Higher Institute for Engineering and Technology, Mansoura, Egypt
| | - Khaled Greish
- Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences Arabian Gulf University, Manama 329, Bahrain.
| |
Collapse
|
6
|
Jiang Y, Wang C, Zu C, Rong X, Yu Q, Jiang J. Synergistic Potential of Nanomedicine in Prostate Cancer Immunotherapy: Breakthroughs and Prospects. Int J Nanomedicine 2024; 19:9459-9486. [PMID: 39371481 PMCID: PMC11456300 DOI: 10.2147/ijn.s466396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Given the global prevalence of prostate cancer in men, it is crucial to explore more effective treatment strategies. Recently, immunotherapy has emerged as a promising cancer treatment due to its unique mechanism of action and potential long-term effectiveness. However, its limited efficacy in prostate cancer has prompted renewed interest in developing strategies to improve immunotherapy outcomes. Nanomedicine offers a novel perspective on cancer treatment with its unique size effects and surface properties. By employing targeted delivery, controlled release, and enhanced immunogenicity, nanoparticles can be synergized with nanomedicine platforms to amplify the effectiveness of immunotherapy in treating prostate cancer. Simultaneously, nanotechnology can address the limitations of immunotherapy and the challenges of immune escape and tumor microenvironment regulation. Additionally, the synergistic effects of combining nanomedicine with other therapies offer promising clinical outcomes. Innovative applications of nanomedicine include smart nanocarriers, stimulus-responsive systems, and precision medicine approaches to overcome translational obstacles in prostate cancer immunotherapy. This review highlights the transformative potential of nanomedicine in enhancing prostate cancer immunotherapy and emphasizes the need for interdisciplinary collaboration to drive research and clinical applications forward.
Collapse
Affiliation(s)
- Yueyao Jiang
- Department of Pharmacy, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Chengran Wang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Chuancheng Zu
- China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Xin’ao Rong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Qian Yu
- Department of Pharmacy, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| | - Jinlan Jiang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China
| |
Collapse
|
7
|
Jiang B, Elkashif A, Coulter JA, Dunne NJ, McCarthy HO. Immunotherapy for HPV negative head and neck squamous cell carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189138. [PMID: 38889878 DOI: 10.1016/j.bbcan.2024.189138] [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: 03/22/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Head and neck cancer (HNSCC) is the 8th most common cancer in the UK, with incidence increasing due to lifestyle factors such as tobacco and alcohol abuse. HNSCC is an immune-suppressive disease characterised by impaired cytokine secretion and dysregulation of immune infiltrate. As such, immunotherapy is a potential treatment option, with therapeutic cancer vaccination demonstrating the greatest potential. The success of cancer vaccination is dependent on informed antigen selection: an ideal antigen must be either tumour-specific or tumour-associated, as well as highly immunogenic. Stratification of the patient population for antigen expression and validated biomarkers are also vital. This review focuses on the latest developments in immunotherapy, specifically the development of therapeutic vaccines, and highlights successes, potential drawbacks and areas for future development. Immunotherapy approaches considered for HNSCC include monoclonal antibodies (mAb), Oncolytic viral (OV) therapies, Immune Checkpoint Inhibitors (ICIs) and cancer vaccines.
Collapse
Affiliation(s)
- Binyumeng Jiang
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Ahmed Elkashif
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Jonathan A Coulter
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
8
|
Sanwick AM, Chaple IF. Targeted radionuclide therapy for head and neck squamous cell carcinoma: a review. Front Oncol 2024; 14:1445191. [PMID: 39239273 PMCID: PMC11374632 DOI: 10.3389/fonc.2024.1445191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a type of head and neck cancer that is aggressive, difficult to treat, and often associated with poor prognosis. HNSCC is the sixth most common cancer worldwide, highlighting the need to develop novel treatments for this disease. The current standard of care for HNSCC usually involves a combination of surgical resection, radiation therapy, and chemotherapy. Chemotherapy is notorious for its detrimental side effects including nausea, fatigue, hair loss, and more. Radiation therapy can be a challenge due to the anatomy of the head and neck area and presence of normal tissues. In addition to the drawbacks of chemotherapy and radiation therapy, high morbidity and mortality rates for HNSCC highlight the urgent need for alternative treatment options. Immunotherapy has recently emerged as a possible treatment option for cancers including HNSCC, in which monoclonal antibodies are used to help the immune system fight disease. Combining monoclonal antibodies approved by the US Food and Drug Administration, such as cetuximab and pembrolizumab, with radiotherapy or platinum-based chemotherapy for patients with locally advanced, recurrent, or metastatic HNSCC is an accepted first-line therapy. Targeted radionuclide therapy can potentially be used in conjunction with the first-line therapy, or as an additional treatment option, to improve patient outcomes and quality of life. Epidermal growth factor receptor is a known molecular target for HNSCC; however, other targets such as human epidermal growth factor receptor 2, human epidermal growth factor receptor 3, programmed cell death protein 1, and programmed death-ligand 1 are emerging molecular targets for the diagnosis and treatment of HNSCC. To develop successful radiopharmaceuticals, it is imperative to first understand the molecular biology of the disease of interest. For cancer, this understanding often means detection and characterization of molecular targets, such as cell surface receptors, that can be used as sensitive targeting agents. The goal of this review article is to explore molecular targets for HNSCC and dissect previously conducted research in nuclear medicine and provide a possible path forward for the development of novel radiopharmaceuticals used in targeted radionuclide therapy for HNSCC, which has been underexplored to date.
Collapse
Affiliation(s)
- Alexis M Sanwick
- Department of Nuclear Engineering, University of Tennessee, Knoxville, TN, United States
| | - Ivis F Chaple
- Department of Nuclear Engineering, University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
9
|
Logotheti S, Pavlopoulou A, Rudsari HK, Galow AM, Kafalı Y, Kyrodimos E, Giotakis AI, Marquardt S, Velalopoulou A, Verginadis II, Koumenis C, Stiewe T, Zoidakis J, Balasingham I, David R, Georgakilas AG. Intercellular pathways of cancer treatment-related cardiotoxicity and their therapeutic implications: the paradigm of radiotherapy. Pharmacol Ther 2024; 260:108670. [PMID: 38823489 DOI: 10.1016/j.pharmthera.2024.108670] [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: 11/11/2023] [Revised: 05/16/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Advances in cancer therapeutics have improved patient survival rates. However, cancer survivors may suffer from adverse events either at the time of therapy or later in life. Cardiovascular diseases (CVD) represent a clinically important, but mechanistically understudied complication, which interfere with the continuation of best-possible care, induce life-threatening risks, and/or lead to long-term morbidity. These concerns are exacerbated by the fact that targeted therapies and immunotherapies are frequently combined with radiotherapy, which induces durable inflammatory and immunogenic responses, thereby providing a fertile ground for the development of CVDs. Stressed and dying irradiated cells produce 'danger' signals including, but not limited to, major histocompatibility complexes, cell-adhesion molecules, proinflammatory cytokines, and damage-associated molecular patterns. These factors activate intercellular signaling pathways which have potentially detrimental effects on the heart tissue homeostasis. Herein, we present the clinical crosstalk between cancer and heart diseases, describe how it is potentiated by cancer therapies, and highlight the multifactorial nature of the underlying mechanisms. We particularly focus on radiotherapy, as a case known to often induce cardiovascular complications even decades after treatment. We provide evidence that the secretome of irradiated tumors entails factors that exert systemic, remote effects on the cardiac tissue, potentially predisposing it to CVDs. We suggest how diverse disciplines can utilize pertinent state-of-the-art methods in feasible experimental workflows, to shed light on the molecular mechanisms of radiotherapy-related cardiotoxicity at the organismal level and untangle the desirable immunogenic properties of cancer therapies from their detrimental effects on heart tissue. Results of such highly collaborative efforts hold promise to be translated to next-generation regimens that maximize tumor control, minimize cardiovascular complications, and support quality of life in cancer survivors.
Collapse
Affiliation(s)
- Stella Logotheti
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece; Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | | | - Anne-Marie Galow
- Institute for Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Yağmur Kafalı
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Efthymios Kyrodimos
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Aris I Giotakis
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Stephan Marquardt
- Institute of Translational Medicine for Health Care Systems, Medical School Berlin, Hochschule Für Gesundheit Und Medizin, 14197 Berlin, Germany
| | - Anastasia Velalopoulou
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, 35043 Marburg, Germany; German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany; Genomics Core Facility, Philipps-University, 35043 Marburg, Germany; Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Jerome Zoidakis
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece; Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, 18057 Rostock, Germany; Department of Life, Light & Matter, Interdisciplinary Faculty, Rostock University, 18059 Rostock, Germany
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece.
| |
Collapse
|
10
|
Iancu D, Fulga A, Vesa D, Zenovia A, Fulga I, Sarbu MI, Tatu AL. Metastatic patterns and treatment options for head and neck cutaneous squamous cell carcinoma (Review). Mol Clin Oncol 2024; 20:40. [PMID: 38756868 PMCID: PMC11097132 DOI: 10.3892/mco.2024.2739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/14/2024] [Indexed: 05/18/2024] Open
Abstract
According to current predictions, one-fifth of all Americans will develop skin cancer during their lifetime. Cutaneous squamous cell carcinoma (cSCC) most commonly occurs in the head and neck region, which is the area of the body with the highest level of sun exposure. High-risk head and neck cSCC (HNcSCC) is a broad category with numerous high-risk factors that are associated with unfavorable results. In cSCC staging systems, clinical and tumor traits that are likely to result in poor outcomes are identified. Metastasis occurs in ~2.5% of patients with cSCC, most often in the local lymph nodes, and there is some indication that lymph node metastasis has a distinct pattern based on the tumor site. Current findings on tumor molecular targets have suggested the use of systemic treatments, particularly immunotherapy (such as cemiplimab, pembrolizumab and nivolumab), over radiotherapy or chemotherapy for this type of metastasis. However, when used simultaneously with immunotherapy, radiotherapy may be beneficial in the treatment of metastatic HNcSCC by improving the efficacy of immunotherapy. The present review aims to assess the existing literature on metastatic HNcSCC pathways and treatment options, in order to define current and future directions. Notably, there is an urgent need to identify patients who may benefit from local or systemic cancer treatments. The treatment of lymph node metastasis presents a therapeutic challenge and requires comprehensive management.
Collapse
Affiliation(s)
- Doriana Iancu
- Department of Otorhinolaryngology, ‘Sfantul Andrei’ Emergency Clinical Hospital of Galati, 800578 Galati, Romania
- Clinical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800010 Galati, Romania
| | - Ana Fulga
- Department of Otorhinolaryngology, ‘Sfantul Andrei’ Emergency Clinical Hospital of Galati, 800578 Galati, Romania
- Clinical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800010 Galati, Romania
| | - Doina Vesa
- Department of Otorhinolaryngology, ‘Sfantul Andrei’ Emergency Clinical Hospital of Galati, 800578 Galati, Romania
- Clinical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800010 Galati, Romania
| | - Andrei Zenovia
- Department of Otorhinolaryngology, ‘Cai Ferate’ General Hospital, 800223 Galati, Romania
| | - Iuliu Fulga
- Clinical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800010 Galati, Romania
- Department of Forensic Medicine, ‘Sfantul Andrei’ Emergency Clinical Hospital of Galati, 800578 Galati, Romania
| | - Mihaela Ionela Sarbu
- Clinical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800010 Galati, Romania
| | - Alin Laurentiu Tatu
- Clinical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800010 Galati, Romania
- Department of Dermatology, ‘Sfanta Cuvioasa Parascheva’ Clinical Hospital of Infectious Diseases, 800179 Galati, Romania
- Multidisciplinary Integrative Center for Dermatologic Interface Research, 800179 Galati, Romania
| |
Collapse
|
11
|
Yang Y, Yang C, Yang Q, Lu S, Liu B, Li D, Li D, Zhang P, Xu P, Lang J, Zhou J. Elucidating Hedgehog pathway's role in HNSCC progression: insights from a 6-gene signature. Sci Rep 2024; 14:4686. [PMID: 38409358 PMCID: PMC10897175 DOI: 10.1038/s41598-024-54937-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/19/2024] [Indexed: 02/28/2024] Open
Abstract
With the emergence of targeted inhibition strategies for Hedgehog signaling in cancer, multiple Hedgehog signaling pathway-related biomarkers have become the focus of research. SsGSEA algorithm was employed to analyze the Hedgehog pathway scores of samples in TCGA-HNSC dataset and divide them into two groups. Weighted co-expression network analysis was performed to identify modules strongly associated with the Hedgehog pathway. Differentially up-regulated genes in tumor samples in comparison to the normal ones were screened by Limma, in which genes belonging to modules strongly related to Hedgehog pathway were further filtered by LASSO reduction and multivariate Cox regression analysis to develop a model. ESTIMATE and CIBERSORT were served to characterize the tumor microenvironment (TME). TIDE assessed immunotherapy response. Hedgehog pathway activity was significantly higher in head and neck squamous cell carcinoma (HNSCC) tissues than in normal tissues and was correlated with HNSCC survival, glycan, cofactors and vitamins, drug metabolism, and matrix scores. Six genes (SLC2A3, EFNB2, OAF, COX4I2, MT2A and TXNRD1) were captured to form a Hedgehog associated 6-gene signature, and the resulting risk score was an independent indicator of HNSCC prognosis. It was significantly positively correlated with stromal score, metabolism, angiogenesis and inflammatory response. Patients in low-risk group with a low TIDE score had higher immunotherapy sensitivity relative to those in high-risk group. This study revealed novel findings of the Hedgehog pathway in HNSCC progression and opened up a Hedgehog pathology-related signature to help identify risk factors contributing to HNSCC progression and help predict immunotherapy outcomes.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oncology, The Third People's Hospital of Chengdu, Chengdu, 610014, China
| | - Chenxi Yang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Qiying Yang
- Military Casualty Management Department, General Hospital of the Western War Zone of the Chinese People's Liberation Army, Chengdu, 610036, China
| | - Shun Lu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Bisheng Liu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Dongyun Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Dongliang Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Peng Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Peng Xu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Jinyi Lang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Jie Zhou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China.
| |
Collapse
|
12
|
Guerrero GG, Minhoto GB, Tibúrcio-Machado CDS, Pinto IAR, Federico CA, Valera MC. Side effects of different head and neck radiotherapy doses on wistar rat's behavior. Brain Res 2024; 1822:148606. [PMID: 37769894 DOI: 10.1016/j.brainres.2023.148606] [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: 06/12/2023] [Revised: 09/12/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Radiotherapy (RT) is a common treatment for head and neck tumors. However, it causes several physical and behavioral side effects, and no study has assessed the emotional effects in rats. Therefore, the present study evaluated the influence of head and neck RT on the behavior and body weight gain in Wistar rats. Fifty-four male Wistar rats were allocated into six groups (n = 9) according to the irradiation dose, which was applied at the first day of the experiment: RT-7.5 (single dose of 7.5 Gy); RT-10 (single dose of 10 Gy); RT-15 (single dose of 15 Gy); RT-30 (single dose of 30 Gy); Control (without RT). The animals were irradiated in the region of the right face, and behavioral tests and weighing were performed on days one, seven, and 28. The open field and Y-maze tests were undertaken to analyze the animal's behavior. The dose of 30 Gy was lethal when applied to the head and neck region. The irradiated animals had less weight gain when compared to the control ones, but there was no statistical difference. In the open field and Y-maze tests, lower mobility of animals in the RT groups was observed both on day seven and at the end of the experiment (day 28) when compared to the control rats (p < 0.05). It was possible to conclude that the different doses of radiation induced depressive behavior in the animals, and that the weight gain tended to be lower in the irradiated groups, however, without statistical difference.
Collapse
Affiliation(s)
- Gustavo Guimarães Guerrero
- Department of Restorative Dentistry, Endodontic Division, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, Brazil
| | - Giovanna Bignoto Minhoto
- Department of Restorative Dentistry, Endodontic Division, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, Brazil
| | - Camilla Dos Santos Tibúrcio-Machado
- Department of Restorative Dentistry, Endodontic Division, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, Brazil
| | - Itza Amarisis Ribeiro Pinto
- Department of Restorative Dentistry, Endodontic Division, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, Brazil
| | | | - Marcia Carneiro Valera
- Department of Restorative Dentistry, Endodontic Division, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, Brazil.
| |
Collapse
|
13
|
Polanowski P, Howorus M, Nasiek A, Kozub A, Pietruszka A, Drosik-Rutowicz K, Polanowska K, Składowski K. Long-Term Survival after Stereotactic Radiotherapy Combined with Immunotherapy in a Patient with Recurrent Oral Cancer. Case Rep Oncol 2024; 17:1366-1373. [PMID: 39650716 PMCID: PMC11623968 DOI: 10.1159/000542321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/25/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Recurrent oral squamous cell carcinoma (SCC) poses significant challenges in treatment, requiring a multifaceted approach for effective management. Case Presentation We present the case of a 68-year-old patient with a history of keratonizing SCC of the mandibular gingiva, treated with surgical resection, adjuvant radiotherapy (RT) to a total dose of 60 Gy in 30 fractions and 6 cycles of concurrent chemotherapy. After 6 years of follow-up, the patient experienced a local late recurrence in clinical stage rT4N0M0 requiring palliative chemotherapy (6 cycles of PF regimen). Due to progression, nivolumab-based immunotherapy was administered. After the 11th cycle of immunotherapy, high-dose re-irradiation (18 Gy in 3 fractions) was applied due to subsequent progression. The addition of stereotactic RT to the immunotherapy allowed nivolumab to be continued until cycle 64, ensuring long-term disease stabilization with acceptable tolerability. Consecutive palliative chemotherapy included paclitaxel and methotrexate. Conclusion This case highlights the complex management of recurrent oral SCC, emphasizing the role of combining stereotactic RT with nivolumab in prolonging the administration of immunotherapy.
Collapse
Affiliation(s)
- Paweł Polanowski
- 1st Radiation and Clinical Oncology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | | | - Aleksandra Nasiek
- 1st Radiation and Clinical Oncology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Anna Kozub
- 3rd Radiation and Clinical Oncology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Agnieszka Pietruszka
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, Cracow, Poland
| | - Katarzyna Drosik-Rutowicz
- 1st Radiation and Clinical Oncology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Katarzyna Polanowska
- Ophthalmology Department, St. Barbara Provincial Hospital No. 5, Sosnowiec, Poland
| | - Krzysztof Składowski
- 1st Radiation and Clinical Oncology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| |
Collapse
|
14
|
Deng H, Wei Z, Du J, Shen Z, Zhou C. Predicting the prognosis, immune response, and immunotherapy in head and neck squamous cell carcinoma using a novel risk model based on anoikis-related lncRNAs. Eur J Med Res 2023; 28:548. [PMID: 38017579 PMCID: PMC10683111 DOI: 10.1186/s40001-023-01521-9] [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: 10/06/2023] [Accepted: 11/09/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is an extremely heterogeneous and metastatic disease. Anoikis, which is a specific type of programmed apoptosis, is involved in tumor metastasis, tissue homeostasis, and development. Herein, we constructed an anoikis-related long non-coding RNA (lncRNA) signature to predict the prognosis, immune responses, and therapeutic effects in HNSCC patients. METHODS A total of 501 HNSCC samples were acquired from the TCGA database and randomly classified into the training and validation groups (1:1 ratio). Thereafter, the results derived from the training set were analyzed with the LASSO regression analysis, and a novel anoikis-related lncRNA risk model was constructed. Time-dependent ROC curves and Kaplan-Meier analysis were carried out to assess the diagnostic value and survival outcomes. A nomogram was utilized to predict the prognostic accuracy. Furthermore, we studied the tumor microenvironment, tumor mutation burden, enrichment pathways, and the response to chemotherapy and immunotherapy. RESULTS Seven anoikis-related lncRNAs (AC015878.1, CYTOR, EMSLR, LINC01503, LINC02084, RAB11B-AS1, Z97200.1) were screened to design a novel risk model, which was recognized as the independent prognostic factor for HNSCC patients. The findings implied that low-risk patients showed significantly longer OS, PFS, and DSS compared to those high-risk patients. The two groups that were classified using the risk model showed significant differences in their immune landscape. The risk model also predicted that low-risk HNSCC patients could attain a better response to immunotherapy, while high-risk patients would be more sensitive to gemcitabine, docetaxel, and cisplatin. CONCLUSIONS We constructed a novel risk model that could be employed for effectively predicting patient prognosis with a good independent prognostic value for HNSCC patients. Furthermore, this model could be used for designing new immunotherapeutic and chemotherapeutic strategies, and it helps clinicians establish personalized and detailed strategies for HNSCC patients.
Collapse
Affiliation(s)
- Hongxia Deng
- Department of Otolaryngology-Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, NingboZhejiang, 315040, China
| | - Zhengyu Wei
- Department of Otolaryngology-Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, NingboZhejiang, 315040, China
| | - Juan Du
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Zhisen Shen
- Department of Otolaryngology-Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, NingboZhejiang, 315040, China
| | - Chongchang Zhou
- Department of Otolaryngology-Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, NingboZhejiang, 315040, China.
| |
Collapse
|
15
|
Mishra VK, Gandhi AK, Rastogi M, Verma R, Khurana R, Hadi R, Sharma V, Agarwal A, Srivastava AK. Retrospective analysis of clinical outcome of 100 inoperable oral cavity carcinoma treated with definitive concurrent chemoradiotherapy with or without induction chemotherapy. Ecancermedicalscience 2023; 17:1630. [PMID: 38414943 PMCID: PMC10898909 DOI: 10.3332/ecancer.2023.1630] [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: 08/20/2023] [Indexed: 02/29/2024] Open
Abstract
Objectives The management of inoperable oral cavity squamous cell carcinoma (OC-SCC) is onerous. We aimed to retrospectively analyse the outcome of our cohort of inoperable OC-SCC treated with definitive concurrent chemoradiotherapy (CTRT) with or without induction chemotherapy (IC). Methods Data of 100 patients (January 2017 to May 2022) of histopathologically proven inoperable OC-SCC treated with definitive CTRT with weekly cisplatin 40 mg/m2 were retrieved from our departmental archives. Radiotherapy (RT) was delivered with three-dimensional conformal plan (66-70 Gy). Toxicities were evaluated using acute morbidity scoring criteria of Radiation Therapy Oncology Group. The response was evaluated as per WHO criteria. Progression free survival (PFS) was calculated from the date of the start of treatment (IC/CTRT) using Kaplan Meier method. Results Median age was 45 years (range 30-80 years). The primary site was oral tongue (59%), retro-molar trigon (15%), buccal mucosa (15%) and others (11%). The stage was III: IVA: IVB in 16:70:14 patients respectively. 72% patients received IC (platinum ± 5 FU ± taxane). Grade 3 skin toxicity, oral mucositis and dysphagia was noted in 13 (13%), 19 (19%) and 13 (13%) patients respectively. The median follow-up duration was 30.5 months (range 6-62 months). Complete response (CR), partial response, progressive disease and death at the time of the last follow-up were 49%, 25%, 15% and 11% respectively. 2-year PFS rate was 49.5%. Stage III patients had a higher CR rate (81.2% versus 42.8%; p = 0.0051) and higher 2-year PFS (81.2% versus 46.4%; p = 0.0056) in comparison to stage IV patients. Conclusion Inoperable patients of OC-SCC treated with definitive CTRT with or without IC yielded CR in approximately half of patients with acceptable toxicity profiles.
Collapse
Affiliation(s)
- Vachaspati Kumar Mishra
- Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Ajeet Kumar Gandhi
- Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Madhup Rastogi
- Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Rakhi Verma
- Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Rohini Khurana
- Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Rahat Hadi
- Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Vikas Sharma
- Department of Surgical Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Akash Agarwal
- Department of Surgical Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Anoop Kumar Srivastava
- Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| |
Collapse
|