Mohsen Nabi Afjadi’s research while affiliated with Tarbiat Modares University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (31)


Risk factors related to head and neck cancers. Head and neck cancers are influenced by several risk factors, with tobacco use (smoking and smokeless forms) and excessive alcohol consumption being the most significant. Prolonged exposure to UV radiation increases the risk of lip cancer, while infection with high-risk HPV, particularly HPV-16, is strongly linked to oropharyngeal cancers. Age plays a role, as these cancers are more common in individuals over 50, and men are at a higher risk than women. A poor diet low in fruits and vegetables may contribute to carcinogenesis, while chronic oral infections and certain bacteria, such as Porphyromonas gingivalis, may also promote malignancy
Generations of CAR-T-cell construct designs. CAR T cells have evolved through five generations, enhancing efficacy and persistence. First-generation CARs include only a CD3ζ signaling domain, while second-generation CARs add a co-stimulatory domain (CD28 or 4-1BB) for better activation. Third-generation CARs incorporate multiple co-stimulatory domains, improving proliferation and cytotoxicity. Fourth-generation CARs (TRUCKs) introduce cytokine genes like IL-12 to modify the tumor microenvironment. Fifth-generation CARs integrate cytokine receptor domains (e.g., IL-2Rβ) for enhanced immune signaling. These advancements improve tumor targeting, durability, and adaptability in immunotherapy
Key challenges in CAR-T cell therapy. CAR-T cell therapy for HNSCCs faces key challenges, including limited infiltration into tumors due to physical barriers in the TME and poor trafficking caused by mismatched chemokine signaling. Tumor heterogeneity further complicates treatment, as diverse TAAs allow cancer cells to evade immune targeting. Additionally, the immunosuppressive TME, dominated by regulatory T cells, myeloid-derived suppressor cells, and inhibitory cytokines like TGF-β and IL-10, diminishes CAR-T cell activity and persistence, significantly limiting therapeutic efficacy
Advances and challenges in CAR-T cell therapy for head and neck squamous cell carcinoma
  • Literature Review
  • Full-text available

May 2025

·

8 Reads

Biomarker Research

Sahand Saeidpour Masouleh

·

·

Ava Ostovar Ravari

·

[...]

·

Mohsen Nabi Afjadi

Head and neck squamous cell carcinoma (HNSCC) remains among the most aggressive malignancies with limited treatment options, especially in recurrent and metastatic cases. Despite advances in surgery, radiotherapy, chemotherapy, and immune checkpoint inhibitors, survival rates remain suboptimal due to tumor heterogeneity, immune evasion, and treatment resistance. In recent years, Chimeric Antigen Receptor (CAR) T-cell therapy has revolutionized hematologic cancer treatment by genetically modifying T cells to target tumor-specific antigens like CD19, CD70, BCMA, EGFR, and HER2, leading to high remission rates. Its success is attributed to precise antigen recognition, sustained immune response, and long-term immunological memory, though challenges like cytokine release syndrome and antigen loss remain. Notably, its translation to solid tumors, including HNSCC, faces significant challenges, such as tumor microenvironment (TME)-induced immunosuppression, antigen heterogeneity, and limited CAR T-cell infiltration. To address these barriers, several tumor-associated antigens (TAAs), including EGFR, HER2 (ErbB2), B7-H3, CD44v6, CD70, CD98, and MUC1, have been identified as potential CAR T-cell targets in HNSCC. Moreover, innovative approaches, such as dual-targeted CAR T-cells, armored CARs, and CRISPR-engineered modifications, aim to enhance efficacy and overcome resistance. Notably, combination therapies integrating CAR T-cells with immune checkpoint inhibitors (e.g., PD-1/CTLA-4 blockade) and TGF-β-resistant CAR T designs are being explored to improve therapeutic outcomes. This review aimed to elucidate the current landscape of CAR T-cell therapy in HNSCC, by exploring its mechanisms, targeted antigens, challenges, emerging strategies, and future therapeutic potential.

Download

Microenvironment-based immunotherapy in oral cancer: a comprehensive review

March 2025

·

18 Reads

·

1 Citation

Medical Oncology

Oral cancer, a prevalent form of head and neck malignancy, accounts for 4% of global cancer cases. The most common type, oral squamous cell carcinoma (OSCC), has a survival rate of about 50%. Even though emerging molecular therapies show promise for managing oral cancer, current treatments like surgery, radiotherapy, and chemotherapy have significant side effects. In addition, the complex tumor microenvironment (TME), involving the extracellular matrix (ECM) and cells like fibroblasts and stromal cells like immune cells, promotes tumor growth and inhibits immune responses, complicating treatment. Nonetheless, immunotherapy is crucial in cancer treatment, especially in oral cancers. Indeed, its effectiveness lies in targeting immune checkpoints such as PD-1 and CTLA-4 inhibitors, as well as monoclonal antibodies like pembrolizumab and cetuximab, adoptive cell transfer methods (including CAR-T cell therapy), cytokine therapy such as IL-2, and tumor vaccines. Thus, these interventions collectively regulate tumor proliferation and metastasis by targeting the TME through autocrine-paracrine signaling pathways. Immunotherapy indeed aims to stimulate the immune system, leveraging both innate and adaptive immunity to counteract cancer cell signals and promote tumor destruction. This review will explore how the TME controls tumor proliferation and metastasis via autocrine-paracrine signaling pathways. It will then detail the effectiveness of immunotherapy in oral cancers, focusing on immune checkpoints, targeted monoclonal antibodies, adoptive cell transfer, cytokine therapy, and tumor vaccines. Graphical abstract Immunotherapy holds a critical position in cancer treatment, particularly in oral cancers. Its effectiveness revolves around immune checkpoints like PD-1 and CTLA-4 inhibitors, targeted monoclonal antibodies such as pembrolizumab and cetuximab, adoptive cell transfer (including CAR-T cell therapy), cytokine therapy like interleukin-2, and tumor vaccines. These interventions collectively target the tumor microenvironment (TME), regulating tumor proliferation and metastasis through autocrine-paracrine signaling pathways.



Fig. 2 P. gingivalis impairs p53 function, a critical tumor suppressor protein. Normally, p53 orchestrates cell responses to stress by inducing cell cycle arrest, promoting DNA repair, initiating apoptosis, or triggering cellular senescence to prevent the development of cancer. P. gingivalis interferes with p53 activity through mechanisms that inhibit its transcriptional and post-translational functions, potentially promoting cell survival and proliferation despite DNA damage or stress. This impairment may contribute to the progression of periodontal disease and increase the risk of oral cancers by disrupting p53-mediated safeguards against tumorigenesis in oral tissues
Exploring the role of oral bacteria in oral cancer: a narrative review

Discover Oncology

A growing body of research indicates that a wide range of cancer types may correlate with human microbiome components. On the other hand, little is known about the potential contribution of the oral microbiota to oral cancer. However, some oral microbiome components can stimulate different tumorigenic processes associated with the development of cancer. In this line, two prevalent oral infections, Porphyromonas gingivalis, and Fusobacterium nucleatum can increase tumor growth. The microbiome can impact the course of the illness through direct interactions with the human body and major modifications to the toxicity and responsiveness to different kinds of cancer therapy. Recent research has demonstrated a relationship between specific phylogenetic groupings and the results of immunotherapy treatment for particular tumor types. Conversely, there has been a recent upsurge in interest in the possibility of using microbes to treat cancer. At the moment, some species, such as Salmonella typhimurium and Clostridium spp., are being explored as possible cancer treatment vectors. Thus, understanding these microbial interactions highlights the importance of maintaining a healthy oral microbiome in preventing oral cancers. From this perspective, this review will discuss the role of the microbiome on oral cancers and their possible application in oral cancer treatment/improvement.


Dormancy of cells and tumor mass dormancy. Circulating tumor cells (CTCs) are tumor cells that have left the main tumor and entered the bloodstream. These aggressive cancer cells, known as DTCs, live in distant organ regions where they self-seed and eventually become dormant. DTCs can either remain dormant for extended periods of time or produce a secondary tumor in a distant organ site either immediately or later (a process known as metastasis). These dormant tumor cells have the potential to awaken from their dormant state and spread throughout time. The cells and tumor mass enter a state of dormancy when the balance between growth and death is reached. The rates of cellular development and death are constrained during dormancy. Autophagy contributes to tumor dormant cell survival and metastasis [21]
Autophagy can cause changes in metabolic processes in cancer cells
The role of mTORC Pathway in autophagy activation during nutrient-rich conditions and nutrient deficiency. By phosphorylating ULK1 and Atg13, mTORC1 inhibits the ULK1 complex. In the PI3KC3 complex I, Atg14, AMBRA1, and NRBF2 are phosphorylated to inhibit the nucleation step of autophagy. MTORC1 inhibits VSP34 activity/LC3 lipidation and the recruitment of phosphatidylinositol phosphates for autophagosome elongation by phosphorylating p300 and WIPI2. By negatively regulating UVRAG and Pacer phosphorylation, mTORC1 inhibits autophagosome-lysosome fusion through PI3KC3 complex II lipid kinase activity and HOPS recruitment. TFEB and TFE3 are relocalized to the nucleus when mTORC1 is inactivated, causing lysosomal and autophagy genes to express. ULK1: Unc-51 Like Autophagy Activating Kinase 1, ATG101: Autophagy-related protein 101, Atg13: Autophagy-related protein 13, Atg14: Autophagy-related protein 14, NRBF2: Nuclear Receptor Binding Factor 2, AMBRA1: Autophagy And Beclin 1 Regulator 1, UVRAG: UV Radiation Resistance Associated gene, PIK3C3: Phosphatidylinositol 3-Kinase Catalytic Subunit Type 3 [71]
Hypoxia-induced autophagy. Hypoxia-inducible factor 1-alpha (HIF-1α) accumulates as a result of hypoxic stress. Targeted by HIF, BNIP3, and BNIP3L are transcriptionally upregulated gene products that compete with the Bcl-XL and Bcl-2 complexes. When BECLIN1 is released from the complex due to competition, multiple autophagic proteins, such as LC3 and ATG5, are recruited, activating the autophagic machinery. BNIP3: Bcl-2 interacting protein 3, Bcl-2: B-cell lymphoma 2, Bcl-XL: B-cell lymphoma-extra-large, LC3: Microtubule-associated Protein 1 Light Chain 3, ATG5: Autophagy protein 5 [84]
Autophagy induced by metabolic processes leads to solid tumor cell metastatic dormancy and recurrence

Medical Oncology

A crucial cellular mechanism that has a complex impact on the biology of cancer, particularly in solid tumors, is autophagy. This review explores how metabolic processes trigger autophagy, which helps metastatic tumor cells go dormant and recur. During metastasis, tumor cells frequently encounter severe stressors, such as low oxygen levels and nutritional deprivation, which causes them to activate autophagy as a survival tactic. This process allows cancer stem cells (CSCs) to withstand severe conditions while also preserving their features. After years of dormancy, dormant disseminated tumor cells (DTCs) may reappear as aggressive metastatic cancers. The capacity of autophagy to promote resistance to treatments and avoid immune detection is intimately related to this phenomenon. According to recent research, autophagy promotes processes, such as the epithelial-to-mesenchymal transition (EMT) and helps build a pre-metastatic niche, which makes treatment strategies more challenging. Autophagy may be a promising therapeutic target because of its dual function as a tumor suppressor in early-stage cancer and a survival promoter in advanced stages. To effectively treat metastatic diseases, it is crucial to comprehend how metabolic processes interact with autophagy and affect tumor behavior. In order to find novel therapeutic approaches that can interfere with these processes and improve patient outcomes, this study highlights the critical need for additional investigation into the mechanisms by which autophagy controls tumor dormancy and recurrence.


CIN, a hallmark of various diseases like cancer, arises from a complex interplay of factors. Epigenetic dysregulation, characterized by changes in gene expression without altering DNA sequences, impacts chromatin structure. Concurrently, centromere erosion and mislocalization of crucial proteins, such as CENP-A, disrupt the precise segregation of chromosomes during cell division. Acentric DNA fragments resulting from chromosome rearrangements may be rescued by the cell, potentially causing instability in subsequent divisions. Neocentromeres, forming at new genomic locations, often exhibit reduced heterochromatin and altered DNA replication, collectively affecting faithful chromosome segregation. Moreover, the mislocalization of key proteins like Aurora B, pivotal in regulating chromosome division, further contributes to the decrease in segregation fidelity. These multifaceted factors intricately intertwine, collectively highlighting the nuanced challenges in maintaining genome stability
Aneuploidy, CIN, and genomic instability are interconnected processes that contribute to the development of tumors. Aneuploidy leads to direct changes in the levels of mRNA and proteins encoded by genes on the abnormal chromosomes. Modifying the dosage of oncogenes and tumor suppressor genes can directly impact cellular transformation. Furthermore, CIN can cause aneuploidy through increased errors in chromosome distribution during cell division, while aneuploidy itself can induce CIN by disrupting the balance of protein complexes necessary for maintaining the genome or by creating imbalances due to the presence of extra DNA. Simultaneously, missegregation of chromosomes has the potential to increase DNA damage and genomic instability. Both CIN and genomic instability are considered mutator phenotypes that heighten the likelihood of accumulating oncogenic mutations, thereby promoting tumor formation. Aneuploidy and DNA damage, which are by-products of these processes, generate genetic variation, enabling cells to have enhanced adaptive potential within the tumor microenvironment
Centromeres in cancer: Unraveling the link between chromosomal instability and tumorigenesis

October 2024

·

68 Reads

·

5 Citations

Medical Oncology

Centromeres are critical structures involved in chromosome segregation, maintaining genomic stability, and facilitating the accurate transmission of genetic information. They are key in coordinating the assembly and help keep the correct structure, location, and function of the kinetochore, a proteinaceous structure vital for ensuring proper chromosome segregation during cell division. Abnormalities in centromere structure can lead to aneuploidy or chromosomal instability, which have been implicated in various diseases, including cancer. Accordingly, abnormalities in centromeres, such as structural rearrangements and dysregulation of centromere-associated proteins, disrupt gene function, leading to uncontrolled cell growth and tumor progression. For instance, altered expression of CENP-A, CENP-E, and others such as BUB1, BUBR1, MAD1, and INCENP, have been shown to ascribe to centromere over-amplification, chromosome missegregation, aneuploidy, and chromosomal instability; this, in turn, can culminate in tumor progression. These centromere abnormalities also promoted tumor heterogeneity by generating genetically diverse cell populations within tumors. Advanced techniques like fluorescence in situ hybridization (FISH) and chromosomal microarray analysis are crucial for detecting centromere abnormalities, enabling accurate cancer classification and tailored treatment strategies. Researchers are exploring strategies to disrupt centromere-associated proteins for targeted cancer therapies. Thus, this review explores centromere abnormalities in cancer, their molecular mechanisms, diagnostic implications, and therapeutic targeting. It aims to advance our understanding of centromeres’ role in cancer and develop advanced diagnostic tools and targeted therapies for improved cancer management and treatment. Graphical abstract Numerical and structural chromosomal instability are genetic alterations that can lead to cancer development. Numerical instability results from abnormal chromosome numbers, while structural instability involves alterations in chromosome structure, disrupting gene function and forming fusion genes. These alterations can lead to genomic instability, promoting uncontrolled cell growth and cancers.


The principle of synthetic lethality. In normal cells, genes A and B collaborate to maintain pathway integrity and ensure cellular functionality, even in the presence of genetic alterations or mutations. If gene A is mutated in cancer cells, it can result in the loss or reduction of its normal activity, which can be detrimental to cell survival. Cancer cells may rely on gene B to sustain pathway activity and support their survival to compensate for this loss. Gene B may either assume some of the functions of gene A or provide alternative pathways to maintain cellular viability. This phenomenon, known as genetic compensation or functional redundancy, highlights the adaptability of cancer cells and their ability to rely on alternative genes for survival. Targeting both gene, A and gene B simultaneously may be crucial in disrupting these compensatory mechanisms and enhancing the effectiveness of cancer treatments. The specific genes involved and their compensatory interactions can vary depending on the cancer type and the specific genetic alterations present in the cells, making it an active area of research in cancer biology
Tools for Identifying SL. Model organisms, direct screening techniques (yeast, medications, RNA interference, and CRISPR technology), and computer analysis (bioinformatics screen) are the sources of the potentially lethal synthetic interaction data. To find out if the synthetic lethal interaction is unique to one or conserved across cell lines, the identified synthetic lethal candidate was verified using panels of human cell lines (Topatana et al. 2020a, b, c)
Targeting DNA Damage Response (DDR) Pathways in cancer therapy. Under normal conditions, the proteins PARP and BRCA1/2 collaborate to repair DNA damage. However, a synthetic lethal (SL) interaction arises when there is a deficiency or inhibition of either the PARP or BRCA1/2 protein. When both the PARP and BRCA1/2 pathways are simultaneously compromised, it results in substantial defects in DNA repair and an accumulation of genetic mutations. This SL interaction exploits the dependence of cancer cells on functional DNA repair mechanisms and offers a potential therapeutic strategy for targeting cancer cells with specific genetic vulnerabilities. Targeting either PARP or BRCA1/2 in cancer cells with pre-existing deficiencies in the other protein disrupts DNA repair processes and promotes the accumulation of lethal DNA damage, ultimately leading to the selective killing of cancer cells. This concept has been successfully applied in the development of PARP inhibitors for the treatment of certain types of cancers, particularly those associated with BRCA1/2 mutations
Potential promising of synthetic lethality in cancer research and treatment

Naunyn-Schmiedeberg's Archives of Pharmacology

Cancer is a complex disease driven by multiple genetic changes, including mutations in oncogenes, tumor suppressor genes, DNA repair genes, and genes involved in cancer metabolism. Synthetic lethality (SL) is a promising approach in cancer research and treatment, where the simultaneous dysfunction of specific genes or pathways causes cell death. By targeting vulnerabilities created by these dysfunctions, SL therapies selectively kill cancer cells while sparing normal cells. SL therapies, such as PARP inhibitors, WEE1 inhibitors, ATR and ATM inhibitors, and DNA-PK inhibitors, offer a distinct approach to cancer treatment compared to conventional targeted therapies. Instead of directly inhibiting specific molecules or pathways, SL therapies exploit genetic or molecular vulnerabilities in cancer cells to induce selective cell death, offering benefits such as targeted therapy, enhanced treatment efficacy, and minimized harm to healthy tissues. SL therapies can be personalized based on each patient’s unique genetic profile and combined with other treatment modalities to potentially achieve synergistic effects. They also broaden the effectiveness of treatment across different cancer types, potentially overcoming drug resistance and improving patient outcomes. This review offers an overview of the current understanding of SL mechanisms, advancements, and challenges, as well as the preclinical and clinical development of SL. It also discusses new directions and opportunities for utilizing SL in targeted therapy for anticancer treatment.


RV exerts anti-inflammatory effects by inhibiting the activation of NF-κB through various mechanisms. Firstly, it blocks the activity of IκB kinase (IKK), preventing the degradation of the inhibitory protein IκB and keeping NF-κB sequestered in the cytoplasm. This prevents NF-κB from translocating to the nucleus and initiating gene transcription related to inflammation. Secondly, RV directly interferes with NF-κB’s DNA-binding activity, disrupting its ability to activate inflammation-related genes. Furthermore, RV can modulate the expression and activity of inhibitory proteins such as IκBα and IκBβ, further inhibiting NF-κB signaling. By acting on multiple levels, RV effectively reduces inflammation by preventing the activation of NF-κB and the subsequent inflammatory response
RV has been found to modulate the MAPK pathway, which is involved in cellular processes, including inflammation. By targeting this pathway, RV may have positive effects on dental health by reducing root resorption and promoting root mineralization in teeth. RV inhibits the activation of key molecules within the MAPK pathway, such as ERK, JNK, and p38 MAPK, which are involved in transmitting inflammatory signals. This inhibition leads to a decrease in the production of pro-inflammatory mediators. By reducing inflammation, RV creates a more favorable environment for preserving the structure of the tooth root. Additionally, inflammation can hinder the natural process of remineralization, but RV’s anti-inflammatory effects support this process by reducing interference and promoting the restoration of mineral content in the root. Thus, RV’s modulation of the MAPK pathway and subsequent reduction in inflammation hold the potential to contribute to the prevention of root resorption and the promotion of remineralization in dental roots
Various factors such as radiation, NDHP oxidase, microbes, nanoparticles, xenobiotics, oxidative stress in mitochondria, ER stress, metabolic reactions in peroxisomes, and enzymatic reactions can contribute to the generation of ROS and oxidative stress, which are associated with dental diseases. However, RV possesses antioxidant properties that can inhibit this process. By scavenging ROS and neutralizing their damaging effects, RV acts as a potent antioxidant. It can directly interact with and neutralize different types of free radicals, while also upregulating the activity and expression of endogenous antioxidant enzymes. This includes enzymes like SOD, catalase, and GPx, which play a crucial role in controlling oxidative stress. Additionally, RV’s anti-inflammatory properties help reduce inflammation, indirectly contributing to its antioxidant actions. Thus, RV, by mitigating oxidative stress, provides protection against dental diseases by preserving the integrity and health of dental tissues
RV inhibits cariogenic bacteria through multiple mechanisms. It disrupts bacterial signaling pathways, particularly quorum sensing, preventing cohesive biofilm formation and reducing pathogenicity. RV also interferes with bacterial adhesion by targeting the expression and function of adhesins, impairing biofilm initiation, and reducing the risk of tooth decay. Additionally, RV disrupts bacterial biofilms by interfering with EPS production and affecting the expression of biofilm-related genes. These actions make the bacteria more susceptible to antimicrobial treatments and inhibit the establishment of persistent infections. Overall, RV shows promise as a therapeutic agent for preventing and managing dental diseases associated with cariogenic bacteria
Promising potential effects of resveratrol on oral and dental health maintenance: a comprehensive review

September 2024

·

79 Reads

·

2 Citations

Naunyn-Schmiedeberg's Archives of Pharmacology

Resveratrol (RV—3, 5, 4′‐trihydroxystilbene) is a natural compound found in plants like red grapes, berries, and peanuts, with promising effects on dental health. It helps strengthen tooth enamel by promoting remineralization, making the teeth more resistant to decay caused by acid-producing bacteria. RV also shields dentin, a vulnerable layer beneath the enamel, from erosion and sensitivity. Its anti-inflammatory properties can reduce inflammation associated with dental conditions such as pulpitis and endodontic diseases. Moreover, RV’s antimicrobial activity inhibits the growth of bacteria involved in dental plaque and biofilm formation, preventing their accumulation on the tooth surface. This contributes to a healthier oral environment and prolongs the lifespan of dental restorative materials. However, the research on RV’s impact on dental health is in its early stages, and further studies are needed to confirm potential benefits. Important factors such as determining the optimal dosage, understanding its bioavailability, and assessing potential side effects require further investigation. This review focuses on the important role of RV in promoting dental health. It delves into various aspects, including its impact on root health, maintenance of the dental pulp, care for tooth enamel, effectiveness of dental restorative materials, and health of dentin. Graphical abstract The effects of resveratrol on dental health.



Mechanisms of PDT. PDT encompasses two main reactions: Type I and Type II. In Type I reactions, the photosensitizer interacts with nearby molecules, forming radical anions or cations, which then react with molecular oxygen to produce ROS like superoxide anions and hydroxyl radicals. These ROS cause oxidative damage to cellular components, including lipids, proteins, and DNA, leading to cellular dysfunction and death. Type II reactions involve the transfer of energy from the excited photosensitizer to ground-state molecular oxygen, producing singlet oxygen, a highly reactive form of oxygen that directly oxidizes biological molecules, causing similar oxidative stress and damage. The combined effects of these reactions create an environment of oxidative stress, overwhelming the cell’s antioxidant defenses, disrupting cellular homeostasis, and triggering cell death pathways such as apoptosis, necrosis, and autophagy. This dual mechanism makes PDT an effective treatment for targeting and destroying cancer cells. ground-state
PDT induces cytotoxicity in target cancer cells through mechanisms involving apoptosis, autophagy, and the modulation of signaling pathways. Apoptosis is driven by caspase activation, mitochondrial dysfunction, and ER stress, which collectively initiate programmed cell death. Additionally, PDT can trigger autophagy, a process where cells degrade and recycle their components, which may lead to cell death if the damage is extensive. Key signaling pathways such as NF-κB/JNK and PI3K/Akt are also influenced by PDT, where their modulation can enhance the apoptotic response. This combination of mechanisms ensures that PDT effectively targets and destroys cancer cells while minimizing damage to surrounding healthy tissue
Histopathological examination of OSCC involves analyzing tissue sections from biopsied or surgically resected specimens. One of the key features evaluated is the differentiation of the tumor. Well-differentiated OSCC closely resembles normal squamous epithelium, exhibiting organized tissue architecture and possible keratinization. Moderately differentiated OSCC shows some loss of differentiation compared to normal cells, with variable sizes and shapes. Poorly differentiated OSCC bears little resemblance to normal squamous cells, lacking organized architecture and often appearing as sheets or cords of cells. Poorly differentiated OSCC tends to grow rapidly and has a higher likelihood of metastasis, resulting in a poorer prognosis compared to well-differentiated and moderately differentiated OSCC. Histopathological examination also considers other features such as tumor invasion patterns, nuclear characteristics, and the presence of an inflammatory infiltrate or desmoplastic reaction. Immunohistochemistry can provide additional information about differentiation, proliferation, and viral etiology, such as HPV infection. Overall, histopathology plays a critical role in diagnosing OSCC, determining its aggressiveness, and guiding treatment decisions
Photodynamic Therapy: A Novel Approach for Head and Neck Cancer Treatment with Focusing on Oral Cavity

August 2024

·

77 Reads

·

6 Citations

Biological Procedures Online

Oral cancers, specifically oral squamous cell carcinoma (OSCC), pose a significant global health challenge, with high incidence and mortality rates. Conventional treatments such as surgery, radiotherapy, and chemotherapy have limited effectiveness and can result in adverse reactions. However, as an alternative, photodynamic therapy (PDT) has emerged as a promising option for treating oral cancers. PDT involves using photosensitizing agents in conjunction with specific light to target and destroy cancer cells selectively. The photosensitizers accumulate in the cancer cells and generate reactive oxygen species (ROS) upon exposure to the activating light, leading to cellular damage and ultimately cell death. PDT offers several advantages, including its non-invasive nature, absence of known long-term side effects when administered correctly, and cost-effectiveness. It can be employed as a primary treatment for early-stage oral cancers or in combination with other therapies for more advanced cases. Nonetheless, it is important to note that PDT is most effective for superficial or localized cancers and may not be suitable for larger or deeply infiltrating tumors. Light sensitivity and temporary side effects may occur but can be managed with appropriate care. Ongoing research endeavors aim to expand the applications of PDT and develop novel photosensitizers to further enhance its efficacy in oral cancer treatment. This review aims to evaluate the effectiveness of PDT in treating oral cancers by analyzing a combination of preclinical and clinical studies. Graphical Abstract


Citations (22)


... The extracellular matrix (ECM), traditionally perceived as a structural entity, has gained recognition as a dynamic regulator of tumor behavior. Composed primarily of collagen, fibronectin, laminins, and proteoglycans, the ECM provides both biochemical and mechanical cues that influence cellular functions such as migration, survival, and drug response [68,83,84]. ...

Reference:

Molecular mechanisms and clinical significance of perineural invasion in malignancies: the pivotal role of tumor-associated Schwann cells in cancer progression and metastasis
Microenvironment-based immunotherapy in oral cancer: a comprehensive review

Medical Oncology

... The gains of 5p, 8p, 19p and losses of 1q, 3p and 17q which similar from several studies [19,[33][34][35]. Abnormalities in chromosomes 1, 3 5, 8, 17 and 19 are described in several cancers such as melanoma, colorectal cancer, neuroblastoma, lung cancer, multiple myeloma, renal cell carcinoma, and oral squamous cell carcinoma (OSCC) [36][37][38][39][40][41][42]. Our findings revealed that the number of CNVs in HCC is higher than in CCA which may reflect greater genomic instability in this cancer that is consistent with its tumor progression, clinical behavior, and aggressiveness [28,35]. ...

Centromeres in cancer: Unraveling the link between chromosomal instability and tumorigenesis

Medical Oncology

... Selain itu, aktivitas antimikroba pada resveratrol menghambat pertumbuhan bakteri yang terlibat dalam plak gigi dan pembentukan biofilm, mencegah akumulasinya pada permukaan gigi. 12 Pada penelitian yang dilakukan oleh Nikniaz, dkk., (2023) terdapat penurunan skor bleeding index yang signifikan yaitu sebanyak 29.10% 4 minggu setelah intervensi.3 Penelitian yang dilakukan oleh Poojitha, dkk., (2024) skor gingival index juga menurun secara signifikan, yang menunjukkan bahwa eliminasi faktor lokal mengurangi peradangan pada jaringan gingiva serta pemberian resveratrol juga meningkatkan efek antiinflamasi.7 Kesimpulannya, resveratrol memiliki sifat antibakteri, antiinflamasi dan dapat melindungi integritas penghalang keratinosit oral yang mencegah infeksi, peradangan dan memfasilitasi regenerasi jaringan. ...

Promising potential effects of resveratrol on oral and dental health maintenance: a comprehensive review

Naunyn-Schmiedeberg's Archives of Pharmacology

... This therapy can induce multiple forms of programmed cell death: apoptosis, necrosis, autophagy, and paraptosis [75,76]. Studies have shown that photodynamic therapy induces apoptosis in cancer cells by activating specific signaling pathways (such as NF-κB/JNK pathway and PI3K/Akt), a process accompanied by cell shrinkage, DNA fragmentation, and formation of apoptotic bodies [77]. Additionally, photodynamic therapy can trigger the intrinsic apoptotic pathway by disrupting mitochondrial function, leading to cytochrome c release and caspase activation; it can also activate the extrinsic pathway through cell surface death receptors, initiating a caspase cascade [77][78][79]. ...

Photodynamic Therapy: A Novel Approach for Head and Neck Cancer Treatment with Focusing on Oral Cavity

Biological Procedures Online

... The anti-inflammatory activity begins in the mouth, with oral administration, from the digestive tract, acting on the conditions that initiate and maintain local inflammation [40]. It is well known that oral lesions accompanied by chronic inflammatory process create an environment that promotes malignant transformation, maintained by the presence of Porphyromonas gingivalis, which is associated with chronic periodontal lesions [41]. In a study using a mucoadhesive film with honey applied directly to oral ulcers in fortyfive Sprague-Dawley rats, a shortening of the healing period of the lesion was observed [42]. ...

The role of periodontitis in cancer development, with a focus on oral cancers

Molecular Biology Reports

... Farhad et al highlighted that polymorphisms in the IL-1 gene cluster, particularly in IL-1β, result in increased cytokine production in response to bacterial invasion. 13 This excessive cytokine release exacerbates inflammation in periodontal tissues, causing collateral damage to the extracellular matrix and alveolar bone. Individuals carrying these polymorphisms are significantly more likely to develop severe forms of periodontal disease, even when exposed to similar microbial challenges as those without such mutations. ...

The role of periodontitis in cancer development, with a focus on oral cancers
  • Citing Article
  • July 2024

Molecular Biology Reports

... Despite breakthroughs in the structure and molecular mechanism of SAA, its underlying aggregation process and pathogenic mechanism remain largely unknown [28]. For years, direct targeted treatments to remove amyloid aggregation and deposition in organs or tissues were not successful, and existing treatments have not been able to reverse or prevent the progression of this disease [29,30]. ...

Therapeutic approaches in proteinopathies
  • Citing Chapter
  • April 2024

Progress in Molecular Biology and Translational Science

... It has been shown that upon subcutaneous administration of the self-assembled GLP-1/glucagon peptide nanofibrils in rodents, the GLP-1/glucagon native peptides were released from the nanofibrils into the blood stream [109]. Self-assembled peptides were significantly considered for numerous advanced functional biomedical applications including bacterial coatings and scaffolds [110,111]. ...

Biotechnological applications of amyloid fibrils
  • Citing Chapter
  • April 2024

Progress in Molecular Biology and Translational Science

... The interaction between laminaran and Dectin-1 activates intracellular signaling cascades, triggering phagocytosis, reactive oxygen species generation, and cytokine release, thereby enhancing innate immune responses. Additionally, laminaran engages Toll-like receptors (TLRs), particularly TLR4, which stimulates the NF-κB signaling pathway, leading to the transcription of pro-inflammatory cytokines [114,115]. Rattigan et al. demonstrated that dietary laminaran supplementation in pigs exposed to dextran sodium sulfate significantly modulated gene expression, including TLR4, MMP2, MMP1, IL6, and IL10 [116]. ...

Potential promising anticancer applications of β-glucans: A review

Bioscience Reports

... AML, as a malignant tumor, still faces many challenges in its treatment [5,7,35]. It has been found that tumor cells can adapt to adverse environments by altering metabolic pathways, and glucose metabolism reprogramming plays a crucial role in this adaptive process [36][37][38]. ...

The role of bone marrow microenvironment (BMM) cells in acute myeloid leukemia (AML) progression: immune checkpoints, metabolic checkpoints, and signaling pathways

Cell Communication and Signaling