This article synthesizes current research on the transcription factor SOX9 as a master regulator of tumor immune evasion and therapy resistance.
This article synthesizes current research on the transcription factor SOX9 as a master regulator of tumor immune evasion and therapy resistance. We explore its dual role in immunobiology, acting as both an oncogene in most cancers and a potential tumor suppressor in specific contexts. The content provides a foundational understanding of SOX9's mechanisms, details methodological approaches for its inhibition, analyzes resistance pathways to conventional immunotherapies like anti-PD-1/anti-LAG-3, and presents comparative evidence validating SOX9 targeting as a strategy to improve patient outcomes. Aimed at cancer researchers and drug development professionals, this review outlines the significant potential of SOX9-directed therapies to overcome limitations of current treatment modalities.
The transcription factor SOX9 (SRY-box transcription factor 9) represents a pivotal regulator of embryonic development and tissue homeostasis whose dysregulation has emerged as a critical contributor to oncogenesis and therapy resistance. As a member of the SOX family characterized by a conserved high mobility group (HMG) box DNA-binding domain, SOX9 recognizes the sequence CCTTGAG and plays essential roles in chondrogenesis, male sex determination, and the development of multiple organs including bone, testis, heart, lung, pancreas, and intestine [1] [2]. Heterozygous mutations in the human SOX9 gene cause campomelic dysplasia, a severe skeletal malformation syndrome frequently accompanied by sex reversal [1]. Beyond its developmental functions, SOX9 has more recently been implicated as a key player in cancer progression, with elevated expression correlated with poor prognosis, metastasis, and treatment resistance across diverse malignancies [3] [4]. This article comprehensively examines the structural domains of SOX9, their functional contributions to both normal physiology and disease processes, and the emerging therapeutic strategies targeting this transcription factor within the broader context of SOX9 inhibition versus conventional immunotherapy outcomes.
The human SOX9 protein comprises 509 amino acids organized into several functionally specialized domains that collectively enable its role as a master transcriptional regulator [1] [4]. Understanding this structural architecture provides the foundation for rational therapeutic design.
Table 1: Functional Domains of Human SOX9 Protein
| Domain | Location | Key Functions | Molecular Interactions |
|---|---|---|---|
| Dimerization Domain (DIM) | N-terminus | Facilitates homo- and heterodimerization | SOXE proteins (SOX8, SOX9, SOX10) |
| HMG Box | Central region | Sequence-specific DNA binding, nuclear localization, DNA bending | Minor groove of DNA (consensus: AGAACAATGG) |
| Transactivation Domain Middle (TAM) | Central region | Synergizes with TAC for transcriptional activation | Transcriptional co-activators |
| Transactivation Domain C-terminal (TAC) | C-terminus | Primary transcriptional activation | MED12, CBP/p300, TIP60, WWP2 |
| PQA-rich Domain | C-terminus | Enhances transactivation capability | Unknown specific partners |
The HMG domain deserves particular emphasis as the defining structural feature of SOX proteins. This domain facilitates sequence-specific DNA binding through minor groove interaction, recognizing the consensus motif AGAACAATGG with AACAAT representing the core binding element [1]. Beyond DNA recognition, the HMG domain contains embedded nuclear localization signals (NLS) and a nuclear export signal (NES) that enable nucleocytoplasmic shuttlingâa dynamic process critical for regulating SOX9 activity [4]. The HMG domain induces significant DNA bending (approximately 70-80°), which likely remodel chromatin architecture and facilitate the assembly of enhanceosome complexes [1].
The dimerization domain enables SOX9 to form both homodimers and heterodimers with other SOXE family members (SOX8 and SOX10). This dimerization capacity is essential for DNA binding and transactivation of specific target genes, particularly in chondrocytes where SOX9 homodimers bind palindromic composite DNA motifs separated by 3-5 nucleotides to activate cartilage-specific genes [1]. Interestingly, SOX9 functions as a monomer in testicular Sertoli cells, demonstrating context-dependent oligomerization [1].
The transactivation domains TAM and TAC synergistically recruit transcriptional co-activators to regulate gene expression. The TAC domain physically interacts with MED12 (mediator complex subunit 12), CBP/p300 (CREB binding protein/E1A binding protein p300), TIP60 (Tat interactive protein-60), and WWP2 (WW domain containing E3 ubiquitin protein ligase 2), thereby enhancing transcriptional activity [1]. This domain is also required for inhibition of β-catenin during chondrocyte differentiation [1]. The less characterized PQA-rich domain further enhances transactivation potential, though it lacks autonomous transactivation capability [1].
Figure 1: Domain Architecture of SOX9 Protein
SOX9 overexpression has been documented in numerous malignancies, including triple-negative breast cancer (TNBC), colorectal cancer, gastric cancer, liver cancer, lung cancer, ovarian cancer, and pancreatic ductal adenocarcinoma [5] [6] [3]. This dysregulation contributes to multiple hallmarks of cancer through diverse molecular mechanisms.
In cancer biology, SOX9 promotes tumor initiation, progression, and metastasis through several interconnected mechanisms. It enhances cancer cell growth, invasion, migration, and metastasis while concurrently fostering therapy resistance [3]. In pancreatic cancer, SOX9 expression maintains cancer stem cell (CSC) populations and promotes invasiveness through nuclear factor-κB (NF-κB) signaling-mediated regulation [7]. Similarly, in hepatocellular carcinoma, SOX9 is necessary for tumor cell initiation, self-renewal, and tumorigenicity in CSCs [7].
The role of SOX9 in therapeutic resistance is particularly noteworthy. Recent research has demonstrated that elevated SOX9 expression contributes to PARP inhibitor (PARPi) resistance in ovarian cancer by enhancing DNA damage repair (DDR) capabilities [8]. SOX9 binds directly to the promoters of key DDR genes (SMARCA4, UIMC1, and SLX4), thereby regulating these critical repair processes [8]. This mechanism represents a novel contributor to treatment failure in ovarian cancer and suggests potential combination therapeutic strategies.
Interestingly, despite its generally oncogenic role, SOX9 demonstrates context-dependent tumor suppressor activity in specific malignancies. A recent study revealed that SOX9 suppresses colon cancer by inhibiting epithelial-mesenchymal transition (EMT) and SOX2 induction [9]. This paradoxical function highlights the complexity of SOX9 biology and underscores the importance of tissue context in determining its functional outcomes.
The compelling evidence linking SOX9 dysregulation to cancer progression and treatment resistance has positioned this transcription factor as an attractive therapeutic target. Several innovative approaches are currently under investigation.
Table 2: Emerging SOX9-Targeted Therapeutic Approaches
| Therapeutic Approach | Mechanism of Action | Development Stage | Key Findings |
|---|---|---|---|
| Multi-epitope peptide vaccine | Induces immune response against SOX9 epitopes | Computational design | High antigenicity predicted; targets B-cell, HTL, and CTL epitopes [5] |
| USP28 inhibition | Promotes SOX9 degradation via ubiquitination | Preclinical | AZ1 inhibitor reduces SOX9 stability, sensitizes ovarian cancer to PARPi [8] |
| Nanocarrier-delivered inhibitors | Targeted delivery of SOX9 inhibitors/siRNA to CSCs | Preclinical concept | Proposed approach to enhance radiotherapy efficacy in GI cancers [7] |
| miRNA-based approaches | Post-transcriptional SOX9 regulation | Preclinical | miR-145 directly targets SOX9, reducing expression in chondrocytes [10] |
The development of a multi-epitope peptide vaccine represents a particularly innovative immunotherapeutic strategy. This vaccine design incorporates B-cell, helper T lymphocyte (HTL), and cytotoxic T lymphocyte (CTL) epitopes with high antigenicity, non-toxicity, and non-allergenicity, linked with appropriate spacers and fused to the 50S ribosomal protein L7/L12 adjuvant [5]. Computational analyses predict that this construct would generate robust cellular and humoral immune responses against SOX9-expressing tumor cells [5].
An alternative strategy involves targeting SOX9 stability through modulation of its protein turnover. Recent research has identified USP28 as a novel interacting partner that inhibits SOX9 ubiquitination and subsequent degradation mediated by the E3 ubiquitin ligase FBXW7 [8]. The USP28-specific inhibitor AZ1 effectively reduces SOX9 protein stability and increases sensitivity of ovarian cancer cells to PARP inhibitors, suggesting promising combination therapeutic potential [8].
Robust experimental models are essential for validating SOX9-targeting strategies. The following methodologies represent key approaches for investigating SOX9 function and therapeutic modulation:
Chromatin Immunoprecipitation Sequencing (ChIP-Seq)
Luciferase Reporter Assays
Co-immunoprecipitation (Co-IP) and Mass Spectrometry
Figure 2: USP28 Inhibition Promotes SOX9 Degradation to Sensitize Cancer Cells to PARP Inhibition
Targeting SOX9 represents a fundamentally different therapeutic approach compared to conventional immunotherapies, with distinct mechanisms of action, limitations, and potential applications.
Conventional cancer immunotherapies, particularly immune checkpoint inhibitors (ICIs) targeting CTLA-4, PD-1, or PD-L1, function by reactivating pre-existing antitumor immunity through blockade of inhibitory pathways in T cells [4]. These approaches have demonstrated remarkable success across multiple cancer types but are limited by variable response rates, immune-related adverse events, and the development of resistance mechanisms.
In contrast, SOX9-targeted approaches address cancer through alternative mechanisms. SOX9 inhibition potentially targets cancer stem cells (CSCs), which are implicated in tumor initiation, metastasis, and therapy resistance [7] [3]. By disrupting this critical cell population, SOX9 targeting may prevent tumor recurrence and overcome treatment resistance. Additionally, SOX9 inhibition may remodel the tumor immune microenvironment. Bioinformatics analyses indicate that SOX9 expression negatively correlates with infiltration of B cells, resting mast cells, resting T cells, monocytes, plasma cells, and eosinophils, while positively correlating with neutrophils, macrophages, activated mast cells, and naive/activated T cells [4]. Thus, SOX9 modulation may potentially convert "cold" tumors into "hot" ones that are more responsive to immunotherapy.
Table 3: Key Research Reagents for SOX9 Investigation
| Reagent/Category | Specific Examples | Research Applications |
|---|---|---|
| SOX9 Antibodies | Anti-SOX9 (AB5535, Sigma-Aldrich); Anti-SOX9 (ab185230, Abcam) | Western blot, immunohistochemistry, immunofluorescence, ChIP |
| Cell Line Models | SKOV3/Ola (PARPi-resistant ovarian cancer); UWB1.289 (ovarian cancer) | Therapy resistance mechanisms, SOX9 functional studies |
| Inhibitors/Modulators | AZ1 (USP28 inhibitor); Olaparib (PARPi) | SOX9 protein stability studies, combination therapy approaches |
| Expression Vectors | SOX9 overexpression constructs; siRNA/shSOX9 vectors | Gain/loss-of-function studies |
| Animal Models | Sox9-deficient mice; Xenograft models | Developmental roles, in vivo therapeutic efficacy |
| Teneligliptin-d8 Carboxylic Acid | Teneligliptin-d8 Carboxylic Acid, MF:C₁₉H₁₇D₈N₅O₂, MW:363.48 | Chemical Reagent |
| Blumenol C glucoside | Blumenol C glucoside, CAS:189109-45-3, MF:C19H32O7 | Chemical Reagent |
SOX9 represents a compelling therapeutic target with complex biology that reflects its dual roles in development and disease. The precise structural domains of SOX9âparticularly the HMG box, dimerization domain, and transactivation domainsâprovide multiple potential interfaces for therapeutic intervention. Current strategies include direct inhibition, protein stability modulation, vaccination approaches, and RNA-based therapies, each with distinct advantages and limitations.
Future research directions should prioritize the development of more specific SOX9 inhibitors with reduced potential for off-target effects on normal developmental processes. The tissue-specific outcomes of SOX9 modulation warrant careful investigation, particularly given its context-dependent tumor suppressor functions in certain malignancies. Additionally, rational combination strategies integrating SOX9 targeting with conventional chemotherapy, radiotherapy, or immunotherapy hold significant promise for overcoming treatment resistance.
As our understanding of SOX9 biology continues to evolve, so too will opportunities for therapeutic intervention. The structural insights and experimental approaches outlined in this review provide a foundation for the continued development of SOX9-targeted therapies that may ultimately improve outcomes for cancer patients facing treatment-resistant disease.
The transcription factor SOX9 (SRY-Box Transcription Factor 9) exemplifies biological duality, acting as a critical regulator in both pathological and physiological processes. As a member of the SOX family featuring a conserved high-mobility group (HMG) box DNA-binding domain, SOX9 plays essential roles in embryonic development, chondrogenesis, and stem cell maintenance [4] [11]. Recent research has illuminated its complex dual role in immunology and disease pathogenesis, functioning as a "double-edged sword" that presents both challenges and opportunities for therapeutic intervention [4] [12]. In cancer, SOX9 frequently acts as an oncogene, promoting tumor initiation, progression, metastasis, and therapy resistance across diverse malignancies [4] [11] [13]. Conversely, in specific physiological contexts, SOX9 contributes beneficially to tissue regeneration and repair, maintaining macrophage function, facilitating cartilage formation, and supporting organ homeostasis [4] [14]. This comparative analysis examines the opposing functions of SOX9 within the broader context of therapeutic targeting, contrasting SOX9 inhibition strategies with conventional immunotherapies.
The dichotomous nature of SOX9 function manifests distinctly across different disease contexts. The table below summarizes key comparative aspects of its pro-tumorigenic versus tissue-repair roles.
Table 1: Comparative Analysis of SOX9 Functions in Pathological vs. Physiological Contexts
| Functional Aspect | Pro-Tumorigenic Role (Cancer Context) | Tissue Repair Role (Physiological Context) |
|---|---|---|
| Primary Function | Promotes tumor progression, metastasis, and therapy resistance [4] [11] [13] | Supports tissue regeneration, cartilage formation, and cellular homeostasis [4] [14] |
| Effect on Immunity | Induces immunosuppression; facilitates immune escape [4] [15] | Maintains macrophage function for tissue repair [4] |
| Therapeutic Implication | Potential target for inhibition in oncology [4] [16] | Potential target for enhancement in degenerative diseases [14] |
| Key Mechanisms | Stemness programming, chemoresistance, immune exclusion [13] [17] | Phagocytic activation, cellular support, maintenance of function [14] |
In cancer, SOX9 drives malignancy through multiple interconnected mechanisms. In High-Grade Serous Ovarian Cancer (HGSOC), SOX9 expression is significantly upregulated following platinum-based chemotherapy and is sufficient to induce a stem-like transcriptional state that confers chemoresistance [13] [17]. SOX9 knockout experiments demonstrate increased platinum sensitivity, while its overexpression promotes significant chemoresistance in vivo [17]. Beyond therapy resistance, SOX9 plays a pivotal role in shaping the immunosuppressive tumor microenvironment. In glioblastoma (GBM), SOX9 expression correlates significantly with immune cell infiltration and checkpoint expression, indicating its involvement in creating an immunosuppressive niche [15]. Similarly, in colorectal cancer, SOX9 expression negatively correlates with anti-tumor immune cells including B cells and resting T cells, while positively correlating with pro-tumor neutrophils and macrophages [4]. SOX9 also contributes to immune evasion by helping latent cancer cells maintain stemness and avoid immune surveillance in secondary metastatic sites [11].
In contrast to its cancer-promoting activities, SOX9 demonstrates protective functions in specific tissue contexts. Recent research in Alzheimer's disease models has revealed that boosting SOX9 levels in astrocytes enhances their ability to clear amyloid plaques, with elevated SOX9 triggering astrocytes to ingest more amyloid deposits and effectively preserve cognitive function [14]. This plaque-clearing activity positions SOX9 as a potential therapeutic target for neurodegenerative disorders. In melanoma development, an intriguing antagonistic relationship exists between SOX9 and the related transcription factor SOX10. Unlike SOX10, which promotes melanoma initiation, SOX9 overexpression induces cell cycle arrest and apoptosis, activating an anti-tumorigenic program that suppresses melanoma formation [18]. Furthermore, in normal tissue homeostasis, SOX9 contributes to cartilage formation and maintenance of macrophage function, supporting its fundamental role in tissue regeneration and repair processes [4].
The dual nature of SOX9 necessitates careful consideration when developing therapeutic strategies. Targeting SOX9 in cancer requires approaches that specifically inhibit its oncogenic functions while preserving or leveraging its beneficial roles.
Several innovative approaches are emerging to target SOX9 for cancer treatment. A novel multi-epitope peptide vaccine targeting SOX9 has been designed for Triple-Negative Breast Cancer (TNBC), incorporating identified B-cell, helper T lymphocyte, and cytotoxic T lymphocyte epitopes with high antigenicity and non-allergenicity [16]. This vaccine construct, fused to the 50S ribosomal protein L7/L12 adjuvant, demonstrates favorable interactions with TLR2 and TLR4 receptors and induces strong cellular and humoral immune responses in simulations [16]. Additionally, research continues to explore direct inhibition strategies aimed at suppressing SOX9 expression or function to counteract its pro-tumorigenic activities, particularly in cancers where it drives stemness and therapy resistance [4] [13].
Conventional immunotherapies, particularly immune checkpoint inhibitors (ICIs), have revolutionized cancer treatment but face limitations that SOX9 targeting may potentially address. The table below compares key characteristics of these approaches.
Table 2: SOX9-Targeted Strategies vs. Conventional Immunotherapies
| Therapeutic Aspect | SOX9-Targeted Approaches | Conventional Immunotherapies (e.g., PD-1/PD-L1 inhibitors) |
|---|---|---|
| Molecular Target | Intracellular transcription factor [16] | Cell surface immune checkpoints [19] |
| Primary Mechanism | Vaccine-induced immune response or direct inhibition [16] | Blocking inhibitory signals on T-cells [19] |
| Therapeutic Challenge | Intracellular location, potential autoimmune reactions [16] | Primary and acquired resistance [19] |
| Resistance Mechanisms | Not fully characterized | Low tumor immunogenicity, impaired T-cell infiltration, immunosuppressive microenvironment [19] |
| Key Applications | TNBC, HGSOC, other SOX9-overexpressing cancers [16] [13] | HNSCC, melanoma, lung cancer, and other solid tumors [19] |
A significant challenge in conventional immunotherapy is PD-1 resistance, particularly in Head and Neck Squamous Cell Carcinoma (HNSCC), where resistance mechanisms include low tumor immunogenicity, impaired T-cell infiltration, and immunosuppressive microenvironments [19]. Research indicates that SOX9, along with Anxa1, plays a role in shaping this tumor-immune standoff, suggesting potential intersections between SOX9 biology and immunotherapy resistance mechanisms [19]. This relationship highlights the promise of combining SOX9-targeted approaches with conventional immunotherapies to overcome resistance and improve treatment outcomes.
Research into SOX9's dual functions employs diverse methodological approaches, from computational immunology to single-cell transcriptomics.
The development of SOX9-targeted vaccines follows a structured computational and experimental workflow:
To investigate SOX9's role in chemotherapy resistance, researchers employ multi-faceted experimental approaches:
Diagram 1: SOX9 signaling pathways in cancer vs. neurodegeneration. SOX9 exhibits context-dependent functions, driving pro-tumorigenic processes in cancer while promoting protective functions in Alzheimer's models.
Research into SOX9's dual functions relies on specialized reagents and experimental tools. The following table catalogizes key resources mentioned across studies.
Table 3: Essential Research Reagents for SOX9 Investigation
| Reagent/Resource | Primary Application | Research Function | Example Use |
|---|---|---|---|
| CRISPR/Cas9 KO | Genetic manipulation | SOX9 knockout to assess functional necessity | Determine chemosensitivity in HGSOC [17] |
| scRNA-Seq | Transcriptomic profiling | Single-cell resolution of SOX9 expression | Analyze tumor heterogeneity pre/post chemotherapy [17] |
| SOX9-Targeting Vaccine | Immunotherapy development | Multi-epitope vaccine construct | Induce immune response against SOX9 in TNBC [16] |
| Anti-SOX9 Antibodies | Protein detection | Immunohistochemistry, Western blot | Validate SOX9 expression in tissue samples [15] [18] |
| Platinum Chemotherapeutics | Chemoresistance studies | Induce SOX9 expression in vitro | Model therapy resistance mechanisms [13] [17] |
| 17-Bromo Vinorelbine Ditartrate | 17-Bromo Vinorelbine Ditartrate | Vinorelbine EP Impurity I | 17-Bromo Vinorelbine Ditartrate is a vinorelbine derivant and a recognized impurity standard for pharmaceutical research. This product is For Research Use Only. Not for diagnostic or human use. | Bench Chemicals |
| 6(7)-Dehydro Fulvestrant-9-sulfone | 6(7)-Dehydro Fulvestrant-9-sulfone | 6(7)-Dehydro Fulvestrant-9-sulfone is a fulvestrant impurity for endocrine therapy research. This product is For Research Use Only. Not for human or veterinary diagnostic or therapeutic use. | Bench Chemicals |
The Janus-faced nature of SOX9 presents both challenges and opportunities for therapeutic development. In cancer contexts, SOX9 drives aggressive tumor phenotypes through stemness programming, therapy resistance, and immune modulation, making it a valuable therapeutic target [4] [13] [17]. Conversely, its protective roles in tissue repair, amyloid clearance, and neural function preservation highlight the importance of context-specific targeting strategies [4] [14]. Future therapeutic approaches must carefully balance the inhibition of SOX9's pro-tumorigenic functions with the preservation of its beneficial physiological roles. Promising strategies include vaccine-based immunotherapies that selectively target SOX9 in malignant cells [16], combination approaches that address SOX9-mediated resistance to conventional treatments [19], and tissue-specific delivery systems that minimize off-target effects. As research continues to elucidate the complex regulatory networks governing SOX9's dual functions, more precise therapeutic interventions will emerge, potentially offering improved outcomes for cancer patients while maintaining essential tissue repair mechanisms.
The SRY-box transcription factor 9 (SOX9) is an evolutionarily conserved transcription factor with a high-mobility group (HMG) DNA-binding domain that recognizes the specific sequence CCTTGAG [20] [2]. Initially identified for its crucial roles in embryonic development, chondrogenesis, and male sex determination [2] [3], SOX9 has more recently emerged as a pivotal regulator of tumor progression and immune modulation. This transcription factor exhibits a complex "dual-key" functionality in immunology, operating as both an oncogenic driver and tumor suppressor in a context-dependent manner [20] [4]. Within the tumor microenvironment, SOX9 significantly influences immune cell infiltration, polarization, and function, thereby shaping anti-tumor immunity and response to conventional immunotherapies [4] [21]. This review synthesizes current evidence positioning SOX9 as a master regulator of immune cell infiltration and function, with particular emphasis on its potential as a therapeutic target whose inhibition may complement or enhance conventional immunotherapy outcomes.
SOX9 demonstrates markedly divergent expression patterns across cancer types, which correspondingly associates with distinct immune infiltration profiles. Comprehensive pan-cancer analyses reveal that SOX9 expression is significantly upregulated in approximately 45% (15 of 33) of cancer types, including colorectal adenocarcinoma (COAD), glioblastoma (GBM), liver hepatocellular carcinoma (LIHC), lung squamous cell carcinoma (LUSC), and pancreatic adenocarcinoma (PAAD) [20]. Conversely, SOX9 expression is decreased in only two cancer types: skin cutaneous melanoma (SKCM) and testicular germ cell tumors (TGCT) [20]. This expression distribution suggests SOX9 primarily functions as a proto-oncogene in most cancer contexts, though it can exhibit tumor-suppressive properties in specific malignancies.
The expression level of SOX9 shows strong correlations with specific immune cell infiltration patterns in the tumor microenvironment, as detailed in Table 1. These correlations were established through bioinformatics analyses of transcriptomic data from The Cancer Genome Atlas (TCGA) and other cohorts, utilizing methods such as gene set enrichment analysis (GSEA) and single-sample GSEA (ssGSEA) [4] [22].
Table 1: Correlation between SOX9 Expression and Immune Cell Infiltration in Human Cancers
| Cancer Type | Immune Correlations with High SOX9 | Prognostic Association |
|---|---|---|
| Colorectal Cancer (CRC) | â B cells, resting mast cells, resting T cells, monocytes, plasma cells, eosinophils [4] | Poor prognosis [3] |
| Lung Adenocarcinoma (LUAD) | â CD8+ T cells, NK cells, dendritic cells [21] | Shorter survival (p=0.0039) [21] |
| Prostate Cancer (PCa) | â CD8+CXCR6+ T cells, activated neutrophils; â Tregs, M2 macrophages [4] | Therapy resistance [3] |
| Glioblastoma (GBM) | Correlation with immune checkpoint expression and specific infiltration patterns [22] | Better prognosis in specific subgroups [22] |
| Multiple Solid Tumors | â CD8+ T cell function, NK cell activity, M1 macrophages; â memory CD4+ T cells [4] | Varies by cancer type |
In lung adenocarcinoma, SOX9 not only suppresses infiltration of cytotoxic immune cells but also elevates collagen-related gene expression and increases collagen fiber deposition [21]. This suggests SOX9 contributes to an immune-suppressive microenvironment by enhancing fibrotic stroma and physical barriers to immune cell penetration.
SOX9 regulates tumor immunity through multiple interconnected mechanistic pathways that modulate both cancer cell-intrinsic properties and the broader tumor microenvironment.
As a transcription factor, SOX9 directly binds to regulatory sequences of target genes to influence immune function. In thymoma, SOX9 expression negatively correlates with genes involved in Th17 cell differentiation, PD-L1 expression, and T-cell receptor signaling pathways [20]. SOX9 can cooperate with c-Maf to activate Rorc and key Tγδ17 effector genes (Il17a and Blk), thereby modulating lineage commitment of early thymic progenitors and influencing the balance between αβ and γδ T-cell differentiation [4].
In prostate cancer, single-cell RNA sequencing and spatial transcriptomics reveal that high SOX9 expression contributes to an "immune desert" phenotype characterized by decreased effector immune cells (CD8+CXCR6+ T cells and activated neutrophils) and increased immunosuppressive cells (Tregs and M2 macrophages) [4]. This landscape effectively excludes cytotoxic immune cells from the tumor core, facilitating immune escape.
In KRAS-driven lung adenocarcinoma, SOX9 significantly elevates collagen-related gene expression and increases collagen fiber deposition [21]. This extracellular matrix remodeling increases tumor stiffness and creates physical barriers that inhibit infiltration of dendritic cells, CD8+ T cells, and NK cells, thereby suppressing anti-tumor immunity independently of direct signaling regulation.
SOX9 expression correlates with immune checkpoint molecule expression in various cancers. In glioblastoma, SOX9 expression shows significant correlation with multiple immune checkpoints, suggesting interconnected regulatory networks [22]. This relationship positions SOX9 within the broader immune checkpoint axis and suggests potential for combination targeting approaches.
The following diagram illustrates the key mechanistic pathways through which SOX9 regulates immune cell infiltration and function in the tumor microenvironment:
Figure 1: SOX9 Regulation of Tumor Immune Microenvironment. SOX9 modulates the tumor immune landscape through three primary mechanisms: (1) Tumor microenvironment remodeling via collagen deposition creating physical barriers to immune cell infiltration; (2) Direct modulation of immune cell signaling pathways affecting differentiation and checkpoint expression; and (3) Tumor-intrinsic changes promoting therapy resistance and epithelial-mesenchymal transition.
The role of SOX9 in immune regulation has been extensively investigated using genetically engineered mouse models (GEMMs). In the KrasG12D-driven lung adenocarcinoma model, researchers utilized both CRISPR/Cas9-mediated Sox9 knockout and Cre-LoxP mediated conditional knockout approaches [21]. The experimental workflow involves intratracheal delivery of Cre recombinase to activate the KrasG12D oncogene while simultaneously deleting Sox9 in specific alleles. These models demonstrated that Sox9 loss significantly reduces lung tumor development, burden, and progression, contributing to longer overall survival [21]. Notably, the tumor-suppressive effects of Sox9 ablation were significantly attenuated in immunocompromised mice compared to syngeneic models, directly implicating the adaptive immune system in SOX9-mediated oncogenesis [21].
Three-dimensional (3D) tumor organoid culture systems have been employed to study SOX9's cell-autonomous functions independent of immune influences. Mouse lung tumor cell lines with low endogenous SOX9 expression (mTC11 and mTC14) were engineered for gain-of-function and loss-of-function studies [21]. These organoid models demonstrated that SOX9 overexpression directly enhances tumor cell proliferation and organoid growth, as quantified by organoid size and cell number per organoid [21].
Comprehensive immune profiling in SOX9 studies employs multiple complementary approaches:
The following diagram illustrates a typical experimental workflow for investigating SOX9-immune interactions using genetic mouse models:
Figure 2: Experimental Workflow for SOX9-Immune Axis Investigation. A typical research pipeline for studying SOX9-immune interactions begins with selection of appropriate genetic models, followed by targeted SOX9 manipulation, monitoring of tumor development parameters, comprehensive immune profiling using multiple analytical platforms, and final validation in different immune contexts.
The emerging understanding of SOX9's role in immune regulation enables direct comparison between SOX9-targeting approaches and conventional immunotherapies, as detailed in Table 2.
Table 2: SOX9 Inhibition Versus Conventional Immunotherapy Mechanisms and Outcomes
| Therapeutic Aspect | SOX9-Targeted Approach | Conventional Immunotherapy |
|---|---|---|
| Primary Mechanism | Transcriptional reprogramming of tumor and immune cells [3] | Blockade of specific immune checkpoints (PD-1, CTLA-4) [4] |
| Target Cell Population | Tumor cells and multiple immune cell subsets [4] [21] | Primarily T-cells [4] |
| Effect on TME | Reduces collagen deposition, decreases physical barriers [21] | Limited effect on physical TME barriers |
| Immune Effects | Increases CD8+ T, NK, and DC infiltration; decreases Tregs, M2 macrophages [4] [21] | Reinvigorates exhausted T-cells [4] |
| Resistance Mechanisms | Emerging but not fully characterized [3] | Well-characterized (T-cell exhaustion, alternative checkpoints) [4] |
| Therapeutic Window | Potential developmental toxicity concerns [2] [3] | Immune-related adverse events [4] |
| Biomarker Status | SOX9 expression levels; correlation with immune exclusion [22] [3] | PD-L1 expression; tumor mutational burden [4] |
SOX9 inhibition represents a fundamentally different therapeutic strategy from conventional immunotherapy by targeting upstream transcriptional master regulators rather than downstream immune checkpoint proteins. While anti-PD-1/PD-L1 antibodies primarily reinvigorate pre-existing but exhausted T-cells, SOX9 targeting addresses the root cause of immune exclusion by modifying the tumor microenvironment to facilitate immune cell infiltration [21]. This includes reduction of collagen deposition and physical barriers, which are not directly addressed by conventional checkpoint inhibitors.
The mechanistic differences between SOX9 inhibition and conventional immunotherapy create compelling rationale for combination approaches. SOX9 targeting may convert "immune-cold" tumors with excluded T-cell patterns into "immune-hot" tumors with infiltrated patterns, thereby increasing the population of patients who could benefit from subsequent checkpoint inhibition [4] [21]. This sequential or simultaneous approach addresses both the physical barriers to immune infiltration (via SOX9 inhibition) and the functional suppression of infiltrated immune cells (via checkpoint blockade).
Table 3: Key Research Reagents and Experimental Models for SOX9-Immune Studies
| Reagent/Model | Specific Example | Research Application |
|---|---|---|
| Cell Lines | 22RV1, PC3, H1975 [20] | In vitro SOX9 manipulation and drug testing |
| Mouse Models | KrasLSL-G12D; Sox9flox/flox GEMM [21] | In vivo tumor-immune interactions |
| Organoid Systems | mTC11, mTC14 3D cultures [21] | Cell-autonomous function studies |
| SOX9 Modulators | Cordycepin (CD) [20] | SOX9 inhibition studies |
| Analytical Tools | HPA, TCGA, GTEx databases [20] [22] | Human expression correlation analysis |
| Antibodies | SOX9 IHC-validated antibodies [20] [21] | Protein expression detection |
| Immune Profiling | Flow cytometry panels (CD8, CD4, NK1.1) [21] | Immune population quantification |
| Direct Red 254 | Direct Red 254, CAS:101380-00-1, MF:C26H24N2O2 | Chemical Reagent |
| μ-Truxilline | μ-Truxilline, CAS:113350-57-5, MF:C10H10N2O | Chemical Reagent |
SOX9 emerges as a master regulator of immune cell infiltration and function within the tumor microenvironment, operating through multiple integrated mechanisms including transcriptional reprogramming, extracellular matrix remodeling, and direct immune cell modulation. The distinct yet complementary mechanisms of SOX9 inhibition compared to conventional immunotherapies present compelling opportunities for novel therapeutic strategies. Specifically, SOX9-targeted approaches may potentially overcome the limitations of current immunotherapies in immune-excluded tumors by addressing the physical and molecular barriers to immune cell infiltration.
Future research directions should focus on developing specific SOX9 inhibitors with favorable therapeutic windows, validating SOX9 as a predictive biomarker for immune-excluded tumors, and designing rational combination trials sequencing or combining SOX9 modulation with established immunotherapies. The continued elucidation of SOX9's complex role in tumor-immune interactions will undoubtedly contribute to more effective immunotherapeutic strategies across multiple cancer types.
The SRY-box transcription factor 9 (SOX9) is a developmental transcription factor crucial for cell differentiation, progenitor cell development, and organogenesis [20] [23]. In recent decades, research has revealed that SOX9 plays a multifaceted role in cancer biology, with its dysregulation implicated in tumor initiation, progression, and therapeutic resistance [3]. This guide provides a comprehensive comparison of the prognostic significance of SOX9 overexpression across various cancer types, synthesizing clinical evidence, experimental data, and molecular mechanisms. The analysis is framed within the broader context of oncological research, particularly exploring the potential of SOX9 inhibition as a therapeutic strategy alongside or in comparison to conventional immunotherapies.
A 2017 meta-analysis of 17 studies encompassing 3,307 patients with solid tumors provided quantitative evidence that SOX9 overexpression significantly correlates with poor survival outcomes [24]. The analysis demonstrated that elevated SOX9 expression was associated with:
This meta-analysis included diverse cancer types including chordoma, osteosarcoma, esophageal cancer, hepatocellular carcinoma, intrahepatic cholangiocarcinoma, pancreatic ductal adenocarcinoma, prostate cancer, thyroid carcinoma, colorectal cancer, gastric cancer, and non-small cell lung cancer [24].
A 2023 study analyzing SOX9 expression across 33 cancer types revealed that SOX9 expression was significantly upregulated in fifteen cancer types compared to matched healthy tissues [20]. The cancers with significantly elevated SOX9 expression included:
Cervical cancer (CESC), Colorectal adenocarcinoma (COAD), Esophageal carcinoma (ESCA), Glioblastoma (GBM), Kidney renal papillary cell carcinoma (KIRP), Lower grade glioma (LGG), Liver hepatocellular carcinoma (LIHC), Lung squamous cell carcinoma (LUSC), Ovarian cancer (OV), Pancreatic adenocarcinoma (PAAD), Rectal adenocarcinoma (READ), Stomach adenocarcinoma (STAD), Thymoma (THYM), Uterine carcinosarcoma (UCS), and Uterine cervical endometrial carcinoma (UCES) [20].
Notably, SOX9 expression was significantly decreased in only two cancers: skin cutaneous melanoma (SKCM) and testicular germ cell tumors (TGCT), suggesting a context-dependent role where SOX9 may occasionally function as a tumor suppressor [20].
Table 1: Prognostic Significance of SOX9 Overexpression Across Specific Cancers
| Cancer Type | Prognostic Impact | Key Statistics | References |
|---|---|---|---|
| Solid Tumors (Overall) | Reduced OS & DFS | OS HR: 1.66 (1.36-2.02); DFS HR: 3.54 (2.29-5.47) | [24] |
| Glioblastoma (GBM) | Diagnostic & prognostic biomarker | Independent prognostic factor for IDH-mutant cases | [15] [22] |
| Liver Hepatocellular Carcinoma (LIHC) | Tumor progression & poor prognosis | Expression features associate with progression | [20] |
| Lung Squamous Cell Carcinoma (LUSC) | Significant upregulation | Increased vs. normal tissue | [20] |
| Pancreatic Adenocarcinoma (PAAD) | Chemoresistance & poor survival | Regulates cancer stem cell characteristics | [3] |
| Breast Cancer | Upregulation in basal-like subtype | Driver of aggressive phenotype | [23] |
| Intrahepatic Cholangiocarcinoma (ICC) | Poor prognosis | Upregulated in cancerous tissues | [25] |
SOX9 promotes tumor progression through multiple interconnected molecular pathways that regulate critical cancer hallmarks:
The SOX9-BMI1-p21CIP axis represents a fundamental mechanism through which SOX9 promotes tumor progression across multiple cancer types, including gastric cancer, glioblastoma, and pancreatic adenocarcinoma [26]. Experimental evidence demonstrates:
This axis enables SOX9 to simultaneously promote proliferation while evading senescence, creating a permissive environment for tumor progression.
SOX9 contributes significantly to treatment resistance through multiple molecular mechanisms:
Recent research has illuminated SOX9's role in resistance to combination immunotherapy. A 2025 study investigating resistance to anti-LAG-3 plus anti-PD-1 therapy in head and neck squamous cell carcinoma identified:
This mechanism represents a novel SOX9-dependent pathway of immunotherapy resistance that operates through modulation of innate immune components.
Research on SOX9 in cancer progression typically employs these key methodological approaches:
1. SOX9 Expression Analysis:
2. Functional Characterization:
3. Mechanism Investigation:
Table 2: Key Research Reagents for SOX9 Investigation
| Reagent/Category | Specific Examples | Research Application | Function in SOX9 Studies |
|---|---|---|---|
| SOX9 Antibodies | Santa Cruz, Abcam, Abnova, Millipore | IHC, Western Blot | SOX9 protein detection and localization |
| Cell Line Models | 22RV1, PC3, H1975, AGS, MKN45, Panc-1, RWP-1, U373, U251 | In vitro functional studies | Investigation of SOX9 roles across cancer types |
| Animal Models | 4NQO-induced HNSCC, Xenograft models, Transgenic mice | In vivo validation | Study of SOX9 in tumor progression and therapy resistance |
| Small Molecule Inhibitors | Cordycepin | SOX9 modulation | Experimental SOX9 inhibition studies |
| Gene Expression Datasets | TCGA, GTEx, GEO databases | Bioinformatic analysis | Correlation of SOX9 with clinical parameters |
| Single-Cell RNA Seq | 10X Genomics Platform | Tumor microenvironment analysis | Identification of SOX9+ cell subpopulations |
| C.I. Acid violet 80 | C.I. Acid violet 80, CAS:12235-17-5, MF:C5H7Cl2OP | Chemical Reagent | Bench Chemicals |
| (3-Hydroxy-1-propenyl)sulfur pentafluoride | (3-Hydroxy-1-propenyl)sulfur pentafluoride, CAS:155990-90-2, MF:C3H5F5OS, MW:184.13 | Chemical Reagent | Bench Chemicals |
Research has identified several promising approaches for targeting SOX9 in cancer:
SOX9 overexpression represents a significant prognostic factor across multiple cancer types, correlating with reduced overall survival, disease-free survival, and more aggressive clinicopathological features. The transcription factor drives tumor progression through diverse mechanisms including regulation of cancer stem cells, promotion of proliferation and survival, induction of therapy resistance, and modulation of immune responses. Emerging evidence particularly highlights SOX9's role in resistance to combination immunotherapies, positioning SOX9 as both a valuable prognostic biomarker and a promising therapeutic target. Future research directions should focus on developing specific SOX9 inhibitors, validating combination approaches with existing therapies, and exploring SOX9's context-dependent roles across different cancer types and molecular subtypes.
The transcription factor SOX9 (SRY-related HMG-box 9) has emerged as a critical regulator in both normal development and oncogenesis. As a member of the SOX family of transcriptional regulators, SOX9 contains a highly conserved high-mobility group (HMG) domain that facilitates DNA binding and influences chromatin architecture [28] [4]. While initially characterized for its essential roles in chondrogenesis, sex determination, and organogenesis, recent evidence has established SOX9 as a key driver in cancer stem cell (CSC) maintenance, tumor initiation, and therapy resistance [28] [17]. CSCs represent a subpopulation within tumors that possess self-renewal capacity, differentiation potential, and enhanced resistance to conventional therapies [28] [29]. This comparative guide synthesizes current experimental evidence defining SOX9's functional roles in CSCs across cancer types, with particular emphasis on its emerging implications for therapeutic targeting in the context of conventional and immunotherapy-resistant malignancies.
SOX9 contributes to cancer stemness through multiple interconnected mechanisms, including maintenance of self-renewal, promotion of therapy resistance, and facilitation of metastatic progression. The table below summarizes key functional roles of SOX9 across different cancer types, based on experimental evidence.
Table 1: Comparative Analysis of SOX9 Functional Roles in Cancer Stem Cells
| Cancer Type | SOX9 Function in CSCs | Experimental Evidence | Clinical/Prognostic Correlation |
|---|---|---|---|
| Hepatocellular Carcinoma (HCC) | CSC marker, self-renewal, differentiation, chemoresistance [30] | SOX9+ cells generated both SOX9+ and SOX9- progeny in vitro and in vivo; formed tumors with as few as 104 cells [30] | Poor recurrence-free survival, stronger venous invasion [30] |
| High-Grade Serous Ovarian Cancer (HGSOC) | Chemoresistance driver, transcriptional reprogramming [17] | Platinum treatment induced SOX9 upregulation within 72 hours; SOX9 knockout increased platinum sensitivity [17] | High SOX9 post-chemotherapy associated with shorter overall survival (HR=1.33) [17] |
| Head and Neck Squamous Cell Carcinoma (HNSCC) | Immunotherapy resistance via immune microenvironment modulation [27] | Sox9+ tumor cells enriched in anti-LAG-3/PD-1 resistant samples; mediated Anxa1-Fpr1 axis impairing neutrophil function [27] | Mediates resistance to combination immunotherapy [27] |
| Lung Cancer | Regulation of CSC properties, metastasis [29] | SOX9 knockdown suppressed sphere formation, ALDH activity, migration, and invasion in SWCNT-transformed cells [29] | Correlates with disease progression and poor survival in NSCLC [29] |
| Multiple Solid Tumors | Promotion of proliferation, senescence inhibition, transformation [28] | Interacts with transcription factors; promotes proliferation, inhibits senescence, collaborates with oncogenes [28] | Overexpression correlates with tumor progression across various cancers [28] |
SOX9 regulates cancer stemness through interaction with key developmental signaling pathways. The diagram below illustrates the primary molecular mechanisms through which SOX9 maintains CSC properties and promotes tumor initiation.
Diagram 1: SOX9-Regulated Signaling in Cancer Stem Cells. SOX9 activates multiple pathways maintaining stemness properties and promoting tumor initiation through self-renewal, differentiation control, and therapy resistance.
Experimental Protocol:
Key Findings: SOX9+ HCC cells demonstrated classical CSC properties, including self-renewal, differentiation into SOX9- progeny, enhanced tumorigenicity (significantly higher frequency at 10â´ cells), and chemoresistance associated with elevated MRP5 expression [30].
Experimental Protocol:
Key Findings: Sox9+ tumor cells drive immunotherapy resistance by transcribing Anxa1, which induces apoptosis of Fpr1+ neutrophils via mitochondrial fission and inhibited mitophagy, ultimately impairing cytotoxic CD8+ T cell and γδT cell infiltration and function [27].
The therapeutic implications of SOX9 targeting versus conventional immunotherapies present distinct mechanistic approaches and potential synergistic opportunities. The table below compares experimental outcomes across intervention strategies.
Table 2: SOX9-Targeted Approaches vs. Conventional Immunotherapy Outcomes
| Therapeutic Approach | Mechanism of Action | Experimental Outcomes | Resistance Mechanisms |
|---|---|---|---|
| SOX9 Inhibition (Genetic ablation) | Direct targeting of CSC self-renewal and maintenance [17] [29] | Increased platinum sensitivity in HGSOC [17]; suppressed metastasis and sphere formation in lung cancer models [29] | Not fully characterized; potential compensatory signaling pathways |
| SOX9 Inhibition (Transcriptional regulation) | Epigenetic modulation of SOX9 expression [17] | Reduced tumor initiation in BCC models [31]; prevented chemoresistance in HGSOC [17] | Chromatin accessibility changes; super-enhancer commissioning [17] |
| Anti-PD-1/PD-L1 Therapy | Immune checkpoint blockade restoring T-cell function [27] | Improved survival in HNSCC models; enhanced tumor immunity [27] | Low tumor immunogenicity; impaired T-cell infiltration; immunosuppressive microenvironment [32] |
| Anti-LAG-3 + Anti-PD-1 Combination | Dual immune checkpoint inhibition [27] | Superior efficacy vs. monotherapy in HNSCC models [27] | Sox9+ tumor cell-mediated neutrophil apoptosis via Anxa1-Fpr1 axis [27] |
| Potential SOX9 Inhibition + Immunotherapy | Targeting CSCs while enhancing immune response | Experimental phase; theoretical synergy in addressing both CSC and immune compartments | Not yet characterized; may prevent Sox9-mediated immunotherapy resistance |
Table 3: Key Research Reagents for SOX9 and Cancer Stem Cell Studies
| Reagent/Category | Specific Examples | Research Application | Experimental Context |
|---|---|---|---|
| SOX9 Reporter Systems | SOX9 promoter-EGFP constructs [30] | FACS isolation of SOX9+ and SOX9- cell populations | HCC CSC isolation and characterization [30] |
| Gene Editing Tools | CRISPR/Cas9 SOX9 knockout [17]; SOX9-targeted shRNAs [29] | Functional validation of SOX9 requirements | Chemosensitivity assays; metastasis studies [17] [29] |
| CSC Functional Assays | Tumor sphere formation; Aldefluor assay; Limiting dilution transplantation [30] [29] | Quantification of stemness properties | Self-renewal capacity; tumor-initiating cell frequency [30] [29] |
| Animal Models | 4NQO-induced HNSCC; Xenograft models; Genetically engineered mice [27] | In vivo therapeutic response assessment | Immunotherapy resistance mechanisms; tumor initiation studies [27] |
| Single-Cell Omics | scRNA-seq; ChIP-seq [17] [27] | Tumor heterogeneity mapping; transcriptional network analysis | SOX9+ subpopulation identification; target gene discovery [17] [27] |
| Immune Profiling Tools | Multiplex immunofluorescence; Flow cytometry panels [27] | Tumor microenvironment characterization | Immune cell infiltration analysis; neutrophil function assays [27] |
| arabino-Hexitol, 3-deoxy-, pentaacetate | arabino-Hexitol, 3-deoxy-, pentaacetate, CAS:134176-59-3, MF:C15H24N4O3 | Chemical Reagent | Bench Chemicals |
| Lys-Pro-AMC | Lys-Pro-AMC, CAS:133066-53-2, MF:C30H29IN2O6 | Chemical Reagent | Bench Chemicals |
The molecular mechanism by which SOX9-expressing tumor cells confer resistance to combination immunotherapy involves a precise signaling axis that disrupts anti-tumor immunity, as illustrated below.
Diagram 2: SOX9-Mediated Immunotherapy Resistance Pathway. SOX9+ tumor cells transcribe ANXA1, which binds FPR1 on neutrophils, inducing mitochondrial dysfunction and apoptosis via BNIP3 downregulation, ultimately impairing cytotoxic T cell infiltration and enabling therapy resistance.
The consolidated evidence positions SOX9 as a master regulator of cancer stemness with multifaceted roles in tumor initiation, therapy resistance, and immune evasion. Experimental data across cancer types demonstrate that SOX9 not only maintains core CSC properties including self-renewal, differentiation, and chemoresistance but also enables immunotherapy resistance through sophisticated modulation of the tumor immune microenvironment. The comparative analysis reveals that while conventional immunotherapies target adaptive immune activation, they remain vulnerable to SOX9-mediated resistance mechanisms, particularly through immunosuppressive neutrophil modulation. Future therapeutic strategies that integrate SOX9 pathway inhibition with immune checkpoint blockade may offer synergistic potential by simultaneously targeting the CSC compartment and restoring anti-tumor immunity, thereby addressing two fundamental pillars of cancer persistence and recurrence.
The transcription factor SOX9 (SRY-related HMG box 9) has emerged as a critical regulator in cancer biology, with dysregulated expression documented across numerous malignancies including ovarian, colorectal, lung, and breast cancers [3] [33]. As a member of the SOX family of transcription factors, SOX9 contains several functional domains: an N-terminal dimerization domain (DIM), a central high-mobility group (HMG) box DNA-binding domain, and C-terminal transcriptional activation domains (TAM and TAC) [4]. This protein structure enables SOX9 to function as a pioneer transcription factor, capable of binding compacted chromatin and initiating transcriptional reprogramming that drives tumor progression, stemness, and therapy resistance [34]. The compelling association between elevated SOX9 expression and poor clinical outcomes has accelerated efforts to develop direct pharmacological inhibitors targeting this oncogenic protein [3] [33].
Table 1: SOX9 Involvement in Human Cancers
| Cancer Type | SOX9's Role | Association with Therapy Resistance | Prognostic Value |
|---|---|---|---|
| Ovarian Cancer | Promotes PARP inhibitor resistance via enhanced DNA damage repair [8] | Stabilizes SOX9 protein; enhances DDR gene expression [8] | Poor survival correlated with high SOX9 [8] |
| Colorectal Cancer | Key blocker of intestinal differentiation; maintains stem cell-like programs [35] | High expression in microsatellite stable (MSS) CRC [35] | Overexpression associated with poor survival [35] |
| Lung Adenocarcinoma | Drives KRAS-induced tumor progression; suppresses anti-tumor immunity [21] | Contributes to EGFR-TKI resistance via Wnt/β-catenin [3] | SOX9-high tumors show significantly shorter survival [21] |
| Breast Cancer | Contributes to tamoxifen resistance; regulates ALDH1A3 and Wnt signaling [8] | Associated with ATP-driven chemoresistance via ABC transporters [8] | Upregulation correlates with poor prognosis [3] |
The development of small molecule inhibitors directly targeting SOX9 represents an active area of investigation, though challenges remain due to the typical difficulty of targeting transcription factors. Researchers at Dana-Farber Cancer Institute have developed specialized assays to support the identification and optimization of SOX9 inhibitors, with preclinical data demonstrating that SOX9 inactivation prevents adenoma formation in established in vivo models [35]. Their approach focuses on disrupting SOX9's ability to block intestinal differentiation in colorectal cancer, thereby promoting cancer cell differentiation and death. This strategy is particularly relevant for microsatellite stable (MSS) colorectal cancer, which represents 85% of CRC cases and is characterized by high SOX9 expression and WNT pathway dependency [35].
An alternative approach to direct SOX9 inhibition involves targeting the regulatory proteins that control SOX9 stability. Recent research has identified USP28 as a key deubiquitinating enzyme that stabilizes SOX9 by inhibiting its FBXW7-mediated ubiquitination and degradation [8]. This mechanism is particularly relevant in ovarian cancer, where the USP28-SOX9 axis promotes resistance to PARP inhibitors like olaparib. Importantly, researchers have demonstrated that the USP28-specific inhibitor AZ1 effectively reduces SOX9 protein stability and sensitizes ovarian cancer cells to PARP inhibitors [8]. This represents a promising indirect strategy for targeting SOX9 function in cancer therapy.
Table 2: Experimental Compounds for SOX9 Pathway Targeting
| Compound/Approach | Molecular Target | Experimental Model | Key Outcomes | Reference |
|---|---|---|---|---|
| USP28 inhibitor AZ1 | USP28 deubiquitinase | Ovarian cancer cell lines (SKOV3, UWB1.289) | Reduces SOX9 protein stability; increases PARPi sensitivity [8] | [8] |
| Dana-Farber SOX9 inhibitor program | SOX9 transcription factor | Colorectal cancer models; intestinal organoids | Promotes differentiation; disrupts SOX9-PROM1 axis; hinders tumor growth [35] | [35] |
| SOX18 inhibitor Sm4 | SOX18 transcription factor (SOXF family) | Zebrafish larvae; mouse breast cancer model | Disrupts SOX18 protein-protein interactions; reduces tumor vascularity and metastasis [36] | [36] |
Cell Line Models: Research into SOX9 targeting typically employs cancer cell lines with documented SOX9 dependency. For ovarian cancer, studies have utilized SKOV3 and UWB1.289 cells, with PARPi-resistant variants (e.g., SKOV3/Ola) generated through progressive olaparib exposure [8]. For colorectal cancer, patient-derived organoids and established CRC lines with high SOX9 expression are preferred for evaluating differentiation-inducing effects of SOX9 inhibitors [35].
Protein Stability Assays: To evaluate compounds targeting SOX9 stability, researchers employ cycloheximide (CHX) chase experiments to measure SOX9 protein half-life, combined with MG132 proteasome inhibition to confirm ubiquitin-mediated degradation pathways [8]. These assays were crucial in demonstrating that USP28 inhibition promotes SOX9 degradation via the ubiquitin-proteasome system.
Three-Dimensional Organoid Cultures: The use of 3D tumor organoid culture systems has proven valuable for assessing SOX9 inhibitors, particularly for evaluating effects on cancer stemness and differentiation. In lung adenocarcinoma models, SOX9-driven tumor organoids show significantly increased size and cell number compared to controls, providing a robust system for inhibitor validation [21].
Genetically Engineered Mouse Models (GEMMs): The KrasLSL-G12D;Sox9flox/flox (KSf/f) mouse model of lung adenocarcinoma has demonstrated that Sox9 loss significantly reduces lung tumor development, burden, and progression, contributing to longer overall survival [21]. Similar models have been employed for colorectal cancer, showing that Sox9 inactivation prevents adenoma formation [35].
Patient-Derived Xenografts (PDX): Engraftment of human tumor tissues into immunocompromised mice allows for evaluation of SOX9 inhibitors on patient-specific cancer contexts. This approach has been particularly useful for studying ovarian cancer models with inherent or acquired PARPi resistance [8].
Syngeneic Graft Models: Comparison of SOX9-driven tumor growth in immunocompetent versus immunocompromised mice has revealed the significant role of SOX9 in modulating the tumor immune microenvironment [21].
SOX9 operates within complex transcriptional networks that govern cell identity and tumor progression. As a pioneer factor, SOX9 can access compacted chromatin regions and initiate transcriptional reprogramming by recruiting histone modifiers and chromatin remodeling complexes [34]. This activity is particularly evident in skin epithelial stem cells, where SOX9 activation redirects epidermal stem cells toward a hair follicle stem cell fate through coordinated enhancement of hair follicle-specific enhancers and silencing of epidermal enhancers.
Diagram 1: SOX9 Regulation and PARPi Resistance Pathway. SOX9 protein stability is controlled by the E3 ubiquitin ligase FBXW7 (promoting degradation) and deubiquitinase USP28 (promoting stabilization). Stable SOX9 translocates to the nucleus and binds promoters of DNA damage repair (DDR) genes, enhancing PARP inhibitor resistance in ovarian cancer [8].
In cancer contexts, SOX9 engages in bidirectional relationships with key signaling pathways. It can be transcriptionally induced by KRAS, NOTCH, EGFR, YAP, NRF2, and TGF-β signaling, while also reinforcing the activity of these pathways through positive feedback loops [21]. In colorectal cancer, SOX9 functions as a critical effector of WNT signaling, maintaining stem cell-like programs that block intestinal differentiation [35]. This positions SOX9 at a convergence point for multiple oncogenic signals, explaining its broad involvement in therapy resistance across cancer types.
The tumor microenvironment modulation by SOX9 presents both challenges and opportunities for cancer therapy. Research in lung adenocarcinoma has revealed that SOX9 suppresses immune cell infiltration by inhibiting tumor-infiltrating CD8+ T cells, natural killer cells, and dendritic cells [21]. This immunosuppressive effect occurs through SOX9-mediated increases in collagen-related gene expression and tumor stiffness, creating a physical barrier to immune cell penetration.
Table 3: SOX9 Inhibition vs. Conventional Immunotherapy
| Parameter | SOX9-Targeted Approach | Conventional Immunotherapy |
|---|---|---|
| Primary Mechanism | Direct targeting of oncogenic transcription factor and its stabilization mechanisms [8] [35] | Immune checkpoint blockade (PD-1/PD-L1, CTLA-4) [37] |
| Effect on Tumor Immunity | Reduces collagen deposition; improves immune cell infiltration [21] | Releases brakes on pre-existing anti-tumor immunity [37] |
| Therapeutic Resistance | Addresses transcription factor-driven stemness and therapy resistance [3] | Addresses immune checkpoint-mediated resistance [37] |
| Cancer Cell-Intrinsic Effects | Promotes differentiation; reverses stemness; inhibits proliferation [35] [34] | Primarily indirect effects on cancer cells via immune activation |
| Biomarker Strategy | SOX9 nuclear expression by IHC; SOX9 transcript levels [35] | PD-L1 expression; tumor mutational burden; MSI status |
When compared to conventional immunotherapy, SOX9 targeting offers complementary mechanisms of action. While immune checkpoint inhibitors work by releasing pre-existing brakes on antitumor immunity, SOX9 inhibition addresses the fundamental transcriptional programming that drives both tumor progression and immune evasion. The combination of these approaches may yield synergistic benefits, particularly in SOX9-high tumors that exhibit excluded immune phenotypes.
Table 4: Key Research Reagents for SOX9 Investigations
| Reagent/Category | Specific Examples | Research Application | Function in SOX9 Studies |
|---|---|---|---|
| Cell Line Models | SKOV3/Ola (PARPi-resistant ovarian cancer); Patient-derived CRC organoids [8] [35] | Drug sensitivity assays; mechanism studies | Provide physiologically relevant systems for SOX9 inhibitor testing |
| Animal Models | KrasLSL-G12D;Sox9flox/flox mice; Patient-derived xenografts [21] | In vivo efficacy studies; tumor microenvironment analysis | Enable assessment of SOX9 targeting in immune-competent contexts |
| Antibodies | Anti-SOX9 (AB5535); Anti-Ki67; Anti-γH2AX; Anti-USP28 [8] | Western blot; IHC; immunofluorescence; Co-IP | Detect SOX9 expression and DNA damage markers; validate target engagement |
| Chemical Inhibitors | USP28 inhibitor AZ1; PARP inhibitor olaparib; Proteasome inhibitor MG132 [8] | Pathway modulation; protein stability assays | Probe SOX9 regulatory mechanisms and combination therapies |
| Molecular Biology Tools | CRISPR/Cas9 Sox9 knockout systems; SOX9 expression vectors [21] | Genetic manipulation; gain/loss-of-function studies | Establish SOX9 dependency; validate specificity of pharmacological inhibitors |
| N-(2-chloroethyl)-4-nitroaniline | N-(2-Chloroethyl)-4-nitroaniline | N-(2-Chloroethyl)-4-nitroaniline (CAS 1965-55-5) is a chemical building block for research. This product is for Research Use Only. Not for human or veterinary use. | Bench Chemicals |
| Odevixibat HCl | Odevixibat HCl|Ileal Bile Acid Transporter (IBAT) Inhibitor | Odevixibat HCl is a potent, selective IBAT inhibitor for research. This product is For Research Use Only (RUO) and is not intended for diagnostic or therapeutic applications. | Bench Chemicals |
Diagram 2: Experimental Workflow for SOX9 Inhibitor Validation. A standardized approach for evaluating direct SOX9 targeting compounds begins with appropriate model selection, progresses through molecular and functional phenotyping, and concludes with comprehensive mechanistic validation [8] [35] [21].
Direct targeting of SOX9 represents a promising frontier in cancer therapeutics, particularly for malignancies characterized by high SOX9 expression and associated therapy resistance. Current approaches include both direct small molecule inhibition and indirect strategies focusing on SOX9 protein stability modulation through regulators like USP28 [8] [35]. The development of robust experimental models and validation methodologies has accelerated progress in this field, though challenges remain in achieving potent and specific SOX9 inhibition.
Future directions will likely focus on combining SOX9-targeted approaches with conventional therapies, including chemotherapy, targeted agents, and immunotherapy. The demonstrated role of SOX9 in modulating the tumor immune microenvironment [21] provides a strong rationale for exploring combinations with immune checkpoint inhibitors, particularly in immune-excluded tumors. Additionally, advances in protein degradation technologies such as PROTACs may offer new avenues for targeting SOX9 through the recruitment of endogenous ubiquitination machinery.
As biomarker strategies evolve, patient selection based on SOX9 nuclear expression or transcript levels will be crucial for clinical development [35]. The integration of SOX9 inhibition into cancer therapeutic regimens holds significant potential for addressing the persistent challenge of therapy resistance across multiple cancer types.
The transcription factor SOX9 (SRY-box transcription factor 9) plays a complex, context-dependent role in oncogenesis and tissue homeostasis. It is frequently overexpressed in diverse malignanciesâincluding glioblastoma, papillary thyroid cancer, ovarian cancer, and othersâwhere it drives proliferation, invasion, chemoresistance, and stem-like properties [38] [17] [39]. Consequently, SOX9 has emerged as a promising therapeutic target. RNA interference (RNAi) technologies, primarily small interfering RNA (siRNA) and microRNA (miRNA) mimics, represent two potent strategies for targeted SOX9 knockdown. This guide objectively compares the performance of these two RNA-based modalities, providing experimental data and protocols to inform therapeutic development within the broader context of SOX9 inhibition and immunotherapy outcomes.
While both siRNA and miRNA operate within the RNAi pathway, their origins, mechanisms of action, and therapeutic profiles differ significantly. The table below summarizes the core characteristics of each molecule.
Table 1: Fundamental Differences Between siRNA and miRNA
| Feature | siRNA (Small Interfering RNA) | miRNA (MicroRNA) |
|---|---|---|
| Origin | Exogenous, synthetic double-stranded RNA [40] | Endogenous, encoded in the genome [41] |
| Biogenesis | Not applicable; chemically synthesized and directly delivered | Processed from pri-miRNA (by Drosha) and pre-miRNA (by Dicer) [41] |
| Complementarity | Perfect or near-perfect complementarity to a single target mRNA sequence [40] [41] | Partial complementarity, primarily via the "seed region" (nucleotides 2-8) [41] |
| Primary Mechanism | mRNA cleavage and degradation via Argonaute 2 (AGO2) [41] | Translational repression and/or mRNA decay [40] [41] |
| Scope of Action | Single-gene targeting; high specificity [40] | Multi-gene regulation; modulates entire networks and pathways [41] |
| Therapeutic Form | Synthetic siRNA duplex [42] | miRNA mimic (duplex resembling mature miRNA) [41] |
The following diagram illustrates the distinct pathways and mechanisms by which synthetic siRNA and miRNA mimics lead to gene silencing.
Direct and indirect targeting of SOX9 using both siRNA and miRNA strategies has demonstrated significant efficacy in preclinical models. The quantitative data from key experiments are summarized below.
Table 2: Experimental Knockdown Efficacy and Functional Outcomes
| Therapeutic Modality | Experimental Model | Key Performance Metrics | Biological Outcome |
|---|---|---|---|
| SOX9-targeting siRNA [38] | Papillary Thyroid Cancer (PTC) Cells (TPC-1, BCPAP) | Significant knockdown at mRNA/protein level (Western Blot, RT-PCR). | Inhibition of proliferation, colony formation, migration, invasion, and EMT; induced apoptosis. |
| miR-101 mimic (targeting SOX9) [39] | Glioblastoma (GBM) Cells (U87MG, U251MG) | ~80-90% reduction in invasion and migration (Transwell assays). | Suppressed proliferation, migration, and invasion in vitro; inhibited tumor growth in vivo. |
| miR-30 family inhibition [43] | Human Intestinal Epithelial Cells (HIECs) | Significant increase in SOX9 mRNA but decrease in SOX9 protein (Western Blot, RT-PCR). | Regulation of SOX9 protein via ubiquitin-proteasome pathway; control of IEC proliferation/differentiation. |
| Engineered amiRNA [44] | Mouse Brain (rAAV9 delivery) | Superior silencing of target gene (Ataxin-2) vs. benchmark amiRNAs (RT-qPCR, IF). | Proof-of-concept for durable in vivo silencing with high processing precision. |
To ensure reproducibility, this section outlines the core methodologies used in the cited studies.
This protocol is adapted from studies in papillary thyroid cancer (PTC) cell lines.
This protocol is adapted from glioblastoma studies using lentiviral delivery of miR-101.
SOX9 operates within key oncogenic signaling networks. Its knockdown exerts therapeutic effects by disrupting these pathways, as illustrated below.
The diagram shows that SOX9 promotes tumorigenesis by maintaining a stem-like transcriptional state [17], activating the Wnt/β-catenin pathway (e.g., in PTC) [38], driving EMT, and inducing chemoresistance [17]. Knockdown of SOX9, therefore, leads to a cascade of therapeutic effects, including inhibited proliferation and invasion, increased apoptosis and differentiation, and restored chemosensitivity. This positions SOX9 inhibition as a strategy to counteract key mechanisms of tumor progression and immune evasion, potentially improving the efficacy of conventional immunotherapies that are often hindered by an immunosuppressive tumor microenvironment and chemoresistance.
Successful implementation of the aforementioned protocols requires the following key reagents and tools.
Table 3: Essential Reagents for SOX9 RNAi Research
| Reagent/Tool | Function/Description | Example Product/Catalog |
|---|---|---|
| SOX9-targeting siRNA | Synthetic duplex for direct, transient SOX9 mRNA knockdown. | Silencer Select Pre-designed siRNA [40] |
| miRNA Mimic (e.g., miR-101) | Synthetic dsRNA that mimics endogenous miRNA to target SOX9 3'UTR. | mirVana miRNA Mimic [40] |
| Lentiviral miR-101 Construct | For stable, long-term expression of miR-101 in cultured cells. | Custom lentiviral particles [39] |
| rAAV9-amiRNA Vector | Recombinant adeno-associated virus serotype 9 for efficient in vivo delivery of artificial miRNAs. | Custom rAAV9 preparation [44] |
| Lipofectamine 2000 | Common transfection reagent for introducing siRNA/mimics into cells. | Thermo Fisher Scientific [38] |
| Anti-SOX9 Antibody | For detecting SOX9 protein levels via Western Blot. | Mouse polyclonal anti-SOX9 (Santa Cruz Biotechnology) [38] |
| SOX9 qRT-PCR Primers | For quantifying SOX9 mRNA expression levels. | Forward: 5â²-TGCTCAAGGGCTACGACTG-3â²Reverse: 5â²-ACGCTTCTCGCTCTCATTCA-3â² [38] |
| 2,5-Diamino-4-imino-1(4H)-naphthalenone | 2,5-Diamino-4-imino-1(4H)-naphthalenone|High-Purity Research Chemical | 2,5-Diamino-4-imino-1(4H)-naphthalenone is a high-purity naphthalenone derivative for research. This product is For Research Use Only. Not for human or veterinary use. |
| 5,6-Dichloro-1,2,3-benzothiadiazole | 5,6-Dichloro-1,2,3-benzothiadiazole|CAS 23620-85-1 |
siRNA and miRNA-mediated knockdown represent two powerful but distinct approaches for targeting SOX9. The choice between them depends on the specific therapeutic goal. siRNA is the preferred tool for achieving potent and specific knockdown of a single gene, making it ideal for validating SOX9's functions and for therapies where precise on-target effects are paramount. In contrast, miRNA mimics (e.g., miR-101) offer a broader, multi-targeted strategy, simultaneously modulating SOX9 and related pathways within its regulatory network. This can be advantageous for treating complex diseases like cancer, where hitting multiple nodes in a pathway can be more effective and reduce the likelihood of resistance.
Future work should focus on optimizing in vivo delivery systems, such as engineered viral vectors [44] or advanced GalNAc conjugates [42], to improve tissue specificity and efficacy. Furthermore, integrating SOX9 knockdown with established immunotherapiesâsuch as immune checkpoint blockadeâpresents a promising frontier for overcoming microenvironment-driven resistance and achieving durable anti-tumor responses.
The transcription factor SOX9 (SRY-related HMG-box 9) plays a complex, context-dependent role in oncogenesis, functioning as both a crucial developmental regulator and a significant driver of tumor progression. As a member of the SOX family, SOX9 contains a highly conserved HMG-box DNA-binding domain that enables recognition of specific DNA sequences and regulation of transcriptional activity [15] [4]. While SOX9 is essential for proper chondrogenesis, bone formation, and organ development, its dysregulation has been implicated in numerous cancer types, including lung adenocarcinoma (LUAD), colorectal cancer (CRC), glioblastoma (GBM), and head and neck squamous cell carcinoma (HNSCC) [15] [27] [21].
In the context of cancer immunotherapy, SOX9 presents a particularly compelling target. Evidence indicates that SOX9 contributes to immunosuppression within the tumor microenvironment (TME) through multiple mechanisms. In KRAS-driven LUAD, SOX9 significantly elevates collagen-related gene expression and increases collagen fibers, potentially increasing tumor stiffness and creating a physical barrier against immune cell infiltration [21]. This remodeling of the extracellular matrix (ECM) suppresses the infiltration and anti-tumor activity of critical immune populations, including CD8+ T cells, natural killer (NK) cells, and dendritic cells [21]. Similarly, in HNSCC, SOX9+ tumor cells drive resistance to combined anti-PD-1 and anti-LAG-3 immunotherapy by directly regulating the expression of annexin A1 (Anxa1), which mediates apoptosis of formyl peptide receptor 1 (Fpr1)+ neutrophils via the Anxa1-Fpr1 axis [27]. This process ultimately impairs the infiltration and tumor-killing capacity of cytotoxic CD8+ T and γδT cells within the TME [27].
The dual nature of SOX9âas both a promoter of tumor progression and a modulator of the immune landscapeâunderscores the therapeutic potential of its inhibition, particularly through advanced nanocarrier systems that can overcome the challenges of conventional drug delivery.
The strategic inhibition of SOX9 in oncology represents an emerging frontier, with biomimetic nano-delivery systems leading current investigative efforts. While multiple studies focus on SOX9 gene delivery for regenerative medicine [45] [46] [47], the targeted inhibition of SOX9 for cancer therapy presents distinct challenges and opportunities. The following analysis synthesizes the current state of SOX9-targeted nanotherapeutics, with a focus on a representative biomimetic platform for SOX9 pathway inhibition.
Table 1: Comparison of SOX9-Related Nanocarrier Systems in Biomedical Research
| Nanocarrier System | Therapeutic Cargo | Target Condition | Primary Objective | Key Findings |
|---|---|---|---|---|
| CMD-BHQ3-PTL/DOX@RBCM [48] | Piceatannol (PTL) & Doxorubicin (DOX) | Colorectal Cancer (CRC) | SOX9 Inhibition via Hippo/YAP1/SOX9 pathway | Effectively suppressed cancer stemness & metastatic potential; pH-responsive release |
| Optimized LNP Platform [45] | SOX5/SOX9 mRNA | Osteoarthritis | SOX9 Activation for cartilage regeneration | Synergistically enhanced ECM synthesis; reduced inflammation |
| Sequentially Assembled Dendrimer [46] | SIRT1 protein & SOX9 plasmid | Intervertebral Disc Degeneration | SOX9 Activation for ECM biosynthesis | Restored mitochondrial homeostasis & ECM metabolism |
| rAAV-sox9/Polymeric Micelles [47] | rAAV-FLAG-hsox9 | Osteoarthritis | SOX9 Activation to counter inflammatory cytokines | Enhanced ECM deposition; counteracted IL-1β & TNF-α effects |
The CMD-BHQ3-PTL/DOX@RBCM system represents a sophisticated approach to targeting SOX9 in colorectal cancer. This biomimetic nanoparticle delivery system was specifically designed to address the challenges of targeting cancer stem cells (CSCs), which are primary drivers of metastasis and are characterized by high SOX9 expression [48].
System Architecture and Components:
Experimental Validation: In vitro studies demonstrated that the formulation effectively inhibited the viability of SW480 and LoVo colorectal cancer cells in a dose-dependent manner, with ICâ â values of 97.44 ± 1.18 µg/mL and 107.33 ± 0.97 µg/mL, respectively [48]. Crucially, the system significantly suppressed CSC markers (CD133, LGR5, CD44) and inhibited tumor cell migration and invasion capabilities, confirming its effectiveness against the metastatic processes driven by SOX9 [48].
The development of effective SOX9-targeted nanocarriers requires rigorous experimental protocols spanning material synthesis, biological validation, and efficacy assessment. The following section outlines key methodological approaches derived from current research.
Table 2: Essential Research Reagents for SOX9-Targeted Nanotherapeutic Development
| Research Reagent/Category | Specific Examples | Function in SOX9 Nanotherapeutic Development |
|---|---|---|
| Lipid Nanoparticle Components | SM-102, DMG-PEG 2000, DSPC, Cholesterol [45] | Form stable, biodegradable vectors for nucleic acid/protein delivery; enhance cellular uptake |
| Polymeric Nanocarriers | Phenylboronic acid-functionalized G5-dendrimer, Carboxymethyl dextran (CMD) [46] [48] | Provide high drug-loading capacity, controlled release kinetics, and targetability |
| Biomimetic Coatings | Red blood cell membrane (RBCM) [48] | Enhance biocompatibility, prolong circulation half-life, reduce immune clearance |
| Characterization Tools | Transmission Electron Microscopy, Dynamic Light Scattering [45] [46] | Assess nanoparticle size, morphology, distribution, and stability |
| SOX9 Pathway Assays | CD133, LGR5, CD44 markers; Hippo/YAP1 components [48] | Validate SOX9 targeting efficacy and elucidate mechanism of action |
Protocol 1: Formulation and Characterization of Biomimetic Nanoparticles
Protocol 2: Evaluation of Anti-CSC Efficacy and SOX9 Pathway Modulation
The development of effective SOX9 inhibitor nanocarriers requires a comprehensive understanding of the molecular pathways regulated by this transcription factor. SOX9 occupies a central position in multiple oncogenic signaling networks, with its inhibition potentially disrupting several key processes that drive tumor progression and therapy resistance.
The following diagram illustrates the pivotal role of SOX9 in cancer progression and the mechanism of a representative nanocarrier system that targets its expression:
Diagram 1: SOX9 in Cancer Signaling and Nanocarrier Inhibition. The Hippo/YAP1/SOX9 pathway drives oncogenic processes, while piceatannol (PTL)-loaded nanocarriers target this axis for therapeutic effect. SOX9 also promotes immune suppression via ANXA1-FPR1 neutrophil interactions [27] [48] [21].
Beyond its direct oncogenic functions, SOX9 plays a multifaceted role in shaping an immunosuppressive tumor microenvironment that limits the effectiveness of conventional immunotherapies. In head and neck squamous cell carcinoma, SOX9+ tumor cells drive resistance to combined anti-PD-1 and anti-LAG-3 therapy through a sophisticated mechanism involving immune cell modulation [27]. Single-cell RNA sequencing analyses of therapy-resistant tumors revealed significant enrichment of SOX9+ tumor cells that directly regulate the expression of annexin A1 (Anxa1) [27].
This SOX9-Anxa1 axis mediates apoptosis of Fpr1+ neutrophils by promoting mitochondrial fission and inhibiting mitophagy through downregulation of BCL2/adenovirus E1B interacting protein 3 (Bnip3) expression [27]. The subsequent reduction of Fpr1+ neutrophils in the TME impairs the infiltration and tumor-killing capacity of cytotoxic CD8+ T and γδT cells, ultimately leading to therapy resistance [27]. This mechanism highlights how SOX9 inhibition could potentially reverse immunotherapy resistance by preventing the depletion of critical immune populations within the tumor microenvironment.
In lung adenocarcinoma, SOX9 contributes to immune evasion through distinct mechanisms, including significant elevation of collagen-related gene expression and increased deposition of collagen fibers within tumors [21]. This ECM remodeling increases tumor stiffness and creates a physical barrier that inhibits the infiltration of anti-tumor immune cells, including CD8+ T cells, natural killer cells, and dendritic cells [21]. The combined effect of these SOX9-mediated immunosuppressive mechanisms underscores the potential of SOX9-targeted nanotherapies to enhance response to conventional immunotherapies by normalizing the tumor immune microenvironment.
The therapeutic targeting of SOX9 via nanocarrier systems presents distinct advantages and potential synergies when compared to, or combined with, established immunotherapeutic approaches. The following analysis contextualizes SOX9 inhibition within the current landscape of cancer immunotherapy.
Table 3: Comparative Analysis of SOX9 Inhibition vs. Conventional Immunotherapy
| Therapeutic Characteristic | SOX9-Targeted Nanotherapy | Immune Checkpoint Inhibition (Anti-PD-1/PD-L1) |
|---|---|---|
| Primary Molecular Target | SOX9 transcription factor & its downstream pathways [48] | PD-1/PD-L1 immune checkpoint proteins [27] |
| Mechanism of Action | Suppression of cancer stemness, metastasis, & immune evasion pathways [48] [21] | Reactivation of exhausted T cells to enhance anti-tumor immunity [27] |
| Therapeutic Resistance | Targets resistant cell populations (CSCs) [48] | Frequently develops via adaptive resistance mechanisms [27] |
| Effect on Tumor Microenvironment | Reduces collagen deposition, inhibits immunosuppressive neutrophil apoptosis [27] [21] | Directly enhances T-cell mediated cytotoxicity [27] |
| Ideal Application Context | Cancers with high SOX9 expression & CSC-driven metastasis (e.g., CRC, HNSCC) [48] [27] | "Immunologically hot" tumors with pre-existing T-cell infiltration [27] |
Emerging evidence suggests that SOX9 inhibition may synergize with conventional immunotherapies to overcome resistance mechanisms. In HNSCC models, SOX9+ tumor cells were significantly enriched in tumors resistant to anti-PD-1 plus anti-LAG-3 combination therapy [27]. This observation suggests that SOX9 inhibition could potentially reverse resistance to dual checkpoint blockade by addressing a non-redundant resistance mechanism. Similarly, in lung adenocarcinoma, SOX9-mediated suppression of dendritic cell infiltration and subsequent inhibition of CD8+ T cell and NK cell activity represents a complementary immunosuppressive pathway to the PD-1/PD-L1 axis [21].
The potential for combination approaches is further supported by the role of SOX9 in regulating multiple aspects of tumor biology. By simultaneously targeting cancer stemness, metastatic potential, and immunomodulation, SOX9 inhibitors delivered via nanocarriers could address multiple resistance mechanisms simultaneously. The CMD-BHQ3-PTL/DOX@RBCM system demonstrates this multi-targeting approach by combining SOX9 pathway inhibition with conventional chemotherapy, resulting in enhanced anti-tumor effects against colorectal cancer models [48].
The development of nanocarrier systems for targeted SOX9 inhibitor delivery represents a promising frontier in oncology therapeutics, particularly for addressing the challenges of cancer stemness, metastasis, and immunotherapy resistance. Current evidence suggests that SOX9 operates as a central node in multiple oncogenic pathways, influencing not only tumor cell intrinsic processes like proliferation and stemness maintenance but also extrinsic factors within the tumor microenvironment that drive immune evasion.
The biomimetic CMD-BHQ3-PTL/DOX@RBCM platform demonstrates the potential of sophisticated nanocarrier systems to simultaneously target SOX9 signaling and deliver conventional chemotherapeutics, resulting in enhanced efficacy against colorectal cancer models [48]. Future directions in this field should focus on expanding the repertoire of SOX9-inhibiting therapeutics, optimizing nanocarrier designs for improved tumor targeting and penetration, and exploring rational combinations with established immunotherapies.
As research advances, SOX9-targeted nanotherapies may offer new hope for patients with aggressive, treatment-resistant malignancies, particularly those characterized by high SOX9 expression and cancer stem cell populations. The integration of SOX9 inhibition into multimodal treatment regimens represents a compelling strategy for addressing the complex, multifaceted nature of cancer progression and therapeutic resistance.
The transcription factor SOX9 (SRY-related HMG-box 9) plays critical roles in development, stem cell regulation, and disease pathogenesis. Emerging evidence positions SOX9 at the nexus of tumor progression, therapy resistance, and immune regulation, making it a compelling therapeutic target. This guide provides an objective comparison of strategies targeting SOX9, with a specific focus on epigenetic modulation, and contrasts these approaches with conventional immunotherapies. We synthesize experimental data and methodologies to equip researchers with practical insights for drug development decisions.
A critical understanding for therapeutic development is SOX9's dual functionality as a pioneer transcription factor. SOX9 can bind to compacted chromatin, initiate nucleosome displacement, and recruit epigenetic modifiers to reshape the transcriptional landscape [34]. This activity enables SOX9 to drive cell fate transitions, including the reprogramming of cells toward stem-like states that underlie therapy resistance in multiple cancers [49] [34].
Table 1: SOX9-Driven Mechanisms of Therapy Resistance Across Cancers
| Cancer Type | SOX9-Linked Resistance Mechanism | Experimental Evidence | Key Regulated Processes |
|---|---|---|---|
| High-Grade Serous Ovarian Cancer | Chemotherapy-induced epigenetic upregulation promotes stem-like state [49] [13]. | CRISPR/Cas9 activation, tumor microarrays, multiomics analysis of patient samples pre/post chemotherapy [49]. | Chemoresistance, self-renewal, continuous proliferation [49]. |
| Head and Neck Squamous Cell Carcinoma (HNSCC) | Mediates resistance to anti-LAG-3 + anti-PD-1 combo therapy via ANXA1-FPR1 axis on neutrophils [27]. | Single-cell RNA-seq in mouse HNSCC model, transgenic mouse validation [27]. | Immune suppression, reduced cytotoxic T and γδ T cell infiltration [27]. |
| High-Grade Glioma (HGG) vs. Ependymoma (EPN) | Context-specific tumor regulation: suppresses HGG growth, promotes EPN progression [50]. | Proteomic analysis in mouse models, manipulation of Sox9 expression [50]. | Tumor-specific protein-protein interactions (histone deacetylase complex vs. oncofusion proteins) [50]. |
The evidence in Table 1 demonstrates that SOX9 mediates resistance through both cell-intrinsic reprogramming and extrinsic modulation of the tumor immune microenvironment. This functional duality necessitates therapeutic strategies that account for its complex biology.
Table 2: Comparison of SOX9-Targeted Approaches vs. Conventional Immunotherapy
| Therapeutic Feature | SOX9-Targeted Strategies (Theoretical/Preclinical) | Conventional Immunotherapy (Checkpoint Inhibitors) |
|---|---|---|
| Primary Target | Master transcription factor and its downstream gene networks [49] [34] [4]. | Immune cell surface receptors (e.g., PD-1, LAG-3) [27] [4]. |
| Mechanism of Action | Inhibition of cancer stem cell reprogramming, chemoresistance, and immune evasion pathways [49] [4]. | Releasing brakes on cytotoxic T cells to restore anti-tumor immunity [27]. |
| Key Challenge | SOX9 is a nuclear protein difficult to target directly; functional heterogeneity across cancers [4] [50]. | Pre-existing or acquired resistance, often mediated by SOX9+ tumor cell subsets [27]. |
| Overcoming Resistance | Targeting downstream effectors (e.g., ANXA1) or epigenetic co-factors [34] [27]. | Combination therapies (e.g., anti-PD-1 + anti-LAG-3), though resistance persists [27]. |
| Therapeutic Scope | Potential to address core mechanisms of tumor initiation, progression, and immune suppression [49] [51]. | Primarily focused on enhancing adaptive immune responses [4] [51]. |
Krt14-rtTA;TRE-Sox9 transgenic mice to reactivate SOX9 in adult epidermal stem cells, triggering fate switching and progression to basal cell carcinoma (BCC)-like lesions [34].
Figure 1: SOX9 pioneer activity drives stemness and therapy resistance. SOX9 binds closed chromatin at target enhancers, recruits epigenetic co-factors to open chromatin, activates a stem-like transcriptional program, and ultimately confers resistance to therapy [49] [34].
Figure 2: SOX9 drives immunotherapy resistance via ANXA1-FPR1 axis. In HNSCC, SOX9+ tumor cells transcribe and secrete ANXA1, which binds FPR1 on neutrophils, inducing their apoptosis via mitochondrial dysfunction. This reduces neutrophil accumulation, impairing cytotoxic T and γδ T cell infiltration and killing, leading to resistance to anti-LAG-3/anti-PD-1 therapy [27].
Table 3: Essential Reagents for Studying SOX9 Biology and Epigenetic Modulation
| Research Reagent | Function/Application | Key Experimental Use |
|---|---|---|
| Doxycycline-Inducible SOX9 Systems | Enables temporal control of SOX9 expression in specific cell types [34]. | Fate-switching experiments in adult epidermal stem cells; studying BCC pathogenesis [34]. |
| CRISPR/Cas9 (Activation/Knockout) | Genetically manipulate SOX9 expression or its downstream targets [49]. | Establishing causal links between SOX9 and chemoresistance; validating effector genes [49] [27]. |
| Anti-SOX9 Antibodies | Detect SOX9 protein (IHC, IF) or map its genomic binding (CUT&RUN, ChIP) [49] [52]. | Identifying SOX9+ cell populations in tumors; epigenomic profiling [49] [34]. |
| 5-Azacytidine | DNA methyltransferase inhibitor; promotes DNA demethylation [52]. | Probing epigenetic regulation of Sox9 expression in chondrocytes [52]. |
| Trimetazidine | Fatty acid oxidation (FAO) inhibitor and AMPK activator [53]. | Investigating SOX9 stability (degradation via reduced phosphorylation) in osteoarthritis models [53]. |
| Anti-ANXA1 / Anti-FPR1 Antibodies | Block the ANXA1-FPR1 interaction [27]. | Testing mechanisms of SOX9-driven immune suppression and therapy resistance [27]. |
| Phloxin | Phloxin, CAS:6441-77-6, MF:C20H4Br4Cl2K2O5, MW:793 g/mol | Chemical Reagent |
| 2-(4-Ethoxyphenyl)-2-methylpropan-1-ol | 2-(4-Ethoxyphenyl)-2-methylpropan-1-ol, CAS:83493-63-4, MF:C12H18O2, MW:194.27 g/mol | Chemical Reagent |
Targeting the epigenetic modulation of SOX9 presents a promising but complex frontier in cancer therapy. The data compiled in this guide underscore that while conventional immunotherapies target immune cell function, SOX9 operates as a master regulator of tumor cell intrinsic resistance and immune evasion. Successful therapeutic strategies will likely require a multi-pronged approach: inhibiting SOX9's pioneer function, disrupting its context-specific protein interactions, or targeting its critical downstream effectors like ANXA1. The choice of model systems and analytical tools, as detailed herein, is critical for deconstructing SOX9's multifaceted roles and translating these insights into novel combination therapies that overcome resistance.
The pursuit of effective cancer therapeutics increasingly focuses on overcoming the formidable challenges of treatment resistance and tumor recurrence. Within this landscape, the transcription factor SOX9 has emerged as a critical regulator of cancer stemness, chemoresistance, and immune evasion across diverse malignancies. This review provides a comprehensive comparison of therapeutic strategies that integrate SOX9 inhibition with conventional chemotherapy and radiotherapy, framing these approaches within the broader context of outcomes research comparing SOX9-targeted interventions with conventional immunotherapies. As research illuminates SOX9's multifaceted roles in tumor biologyâfrom maintaining cancer stem cells to sculpting immunosuppressive microenvironmentsâthe strategic value of targeting SOX9 alongside established treatment modalities becomes increasingly evident. This analysis synthesizes current experimental evidence, detailed methodologies, and quantitative outcomes to guide researchers and drug development professionals in advancing this promising therapeutic paradigm.
SOX9 (SRY-Box Transcription Factor 9) is a nuclear transcription factor belonging to the SOX family, characterized by a highly conserved high-mobility group (HMG) box domain that facilitates DNA binding and transcriptional regulation [15] [4]. The protein structure includes several functional domains: an N-terminal dimerization domain (DIM), the central HMG box domain responsible for DNA recognition and nuclear localization, and two transcriptional activation domains (TAM and TAC) at the C-terminus that interact with various cofactors to enhance transcriptional activity [4]. This structural configuration enables SOX9 to function as a master regulator of developmental processes, stem cell maintenance, and tissue homeostasis.
In oncological contexts, SOX9 undergoes pathological dysregulation, driving multiple hallmarks of cancer. It is frequently overexpressed in diverse malignancies including glioblastoma (GBM), ovarian cancer, breast cancer, colorectal cancer, and head and neck squamous cell carcinoma [15] [54] [11]. SOX9 expression correlates strongly with advanced tumor grade, metastatic progression, and poor survival outcomes across cancer types [4] [11]. Mechanistically, SOX9 promotes tumor initiation and propagation by enriching cancer stem cell (CSC) populationsâself-renewing, therapy-resistant cells that drive tumor recurrence [54]. In ovarian cancer, SOX9 expression is significantly upregulated in chemoresistant cells, where it functions as a master regulator reprogramming cancer cells into stem-like, tumor-initiating cells capable of continuous self-renewal and proliferation [54]. Beyond stemness regulation, SOX9 facilitates immune evasion by impairing immune cell function and fostering an immunosuppressive tumor microenvironment [4].
Table 1: SOX9 Dysregulation Across Cancer Types
| Cancer Type | Expression Pattern | Functional Consequences | Prognostic Impact |
|---|---|---|---|
| Glioblastoma (GBM) | Highly expressed in tumor tissues [15] | Promotes immune infiltration & checkpoint expression [15] | Better prognosis in lymphoid invasion subgroups [15] |
| Ovarian Cancer | Upregulated in chemoresistant cells [54] | Reprograms cancer cells into stem-like cells [54] | Contributes to chemotherapy resistance [54] |
| Breast Cancer | Frequently overexpressed [11] | Regulates tumor initiation, proliferation, migration [11] | Associated with aggressive basal-like subtype [11] |
| Colorectal Cancer | Highly expressed in CRC tissues [55] | Promotes stemness, metastasis [55] | Independent poor prognostic factor [55] |
| Head and Neck Squamous Cell Carcinoma | Enriched in therapy-resistant samples [27] | Regulates ANXA1-FPR1 axis to suppress immune cell function [27] | Mediates resistance to combination immunotherapy [27] |
SOX9 drives chemotherapy resistance through multiple interconnected mechanisms. In ovarian cancer, SOX9 is epigenetically upregulated in response to chemotherapy treatment, as demonstrated in both cell lines and patient samples collected before and after chemotherapy [54]. This upregulation promotes a stem-cell-like phenotype characterized by enhanced self-renewal capacity and reduced apoptosis. Similarly, in glioblastoma, SOX9 expression is associated with temozolomide (TMZ) resistance, where it functions as a super-enhancer-associated gene that maintains tumor cell survival under therapeutic pressure [56]. The stemness-promoting function of SOX9 enables a subpopulation of therapy-resistant cells to survive initial treatment and eventually drive disease recurrence.
Recent investigations have demonstrated that targeting SOX9 can reverse chemoresistance and synergize with conventional chemotherapeutic agents. In GBM, the super-enhancer inhibitor THZ2 (targeting CDK7) suppressed SOX9 expression and reversed TMZ resistance [56]. Both THZ2 and JQ1 (a BRD4 inhibitor) exhibited synergistic antitumor effects when combined with TMZ in GBM cells, with combination indices indicating enhanced efficacy beyond either approach alone [56].
Table 2: Experimental Models of SOX9 Inhibition with Chemotherapy
| Cancer Model | SOX9-Targeting Agent | Chemotherapeutic Agent | Key Findings | Experimental Methods |
|---|---|---|---|---|
| Glioblastoma (GBM) cells | THZ2 (CDK7 inhibitor) [56] | Temozolomide (TMZ) [56] | Synergistic antitumor effects; Reversal of TMZ resistance [56] | Cell viability (CCK-8), colony formation, CUT&RUN assays [56] |
| Glioblastoma (GBM) cells | JQ1 (BRD4 inhibitor) [56] | Temozolomide (TMZ) [56] | Synergistic antitumor effects [56] | Cell viability (CCK-8), combination index calculation [56] |
| Ovarian cancer cells | CRISPR/Cas9 SOX9 knockout [54] | Conventional chemotherapy (unspecified) [54] | Reduced chemoresistance; Decreased stem-like properties [54] | CRISPR/Cas9 gene editing, transcriptome analysis, single-cell RNA sequencing [54] |
| Colorectal cancer cells | Evodiamine (USP4/SOX9 axis inhibitor) [55] | Not combined with chemo but showed reduced stemness [55] | Inhibited stemness; SOX9 protein destabilization [55] | Ubiquitination assays, Western blot, qPCR, colony formation [55] |
For investigating SOX9 inhibition in combination with chemotherapy, several well-established experimental approaches have been employed:
Establishment of Chemoresistant Cell Lines: Log-phase cancer cells are seeded in 96-well plates and exposed to progressively increasing concentrations of chemotherapeutic agents. For instance, in GBM studies, U87MG cells were initially exposed to TMZ at 1/100 of the IC50 (0.0121 mM), with stepwise increases (0.02, 0.04, 0.08, 0.16, 0.32, 0.64, 1.0 mM) upon cellular adaptation. Each concentration was maintained for 14 days before escalation to establish stable resistant lineages [56].
Cell Viability and Combination Assays: Cell viability is assessed using CCK-8 assays. Cells are seeded in 96-well plates at optimized densities (5 à 10³ cells/well for standard assays or 2 à 10³ cells/well for time-course studies) and treated with gradient concentrations of SOX9-targeting agents and chemotherapeutics. After specified time points, CCK-8 solution is added and incubated for 1 hour at 37°C before measuring absorbance at 450nm. Combination indices are calculated using the Chou-Talalay method to quantify synergistic effects [56].
Molecular Analyses of SOX9 Function: CUT&RUN assays examine protein-DNA interactions to identify SOX9 binding sites. Cells are harvested, bound to concanavalin A-coated beads, and incubated with antibodies against SOX9, CDK7, or BRD4. Protein A-MNase is added to cleave bound DNA, which is then extracted and sequenced. Additionally, Western blotting validates SOX9 protein expression using specific antibodies, while qPCR measures transcriptional changes in stemness markers like OCT4 and CD133 [56] [55].
While direct evidence linking SOX9 to radiotherapy resistance is more limited in the available literature, compelling indirect evidence suggests a potential role. SOX9 is a well-established regulator of cancer stem cells (CSCs) [54] [11], and CSCs are known to contribute significantly to radioresistance across multiple cancer types. These stem-like cells exhibit enhanced DNA repair capacity, activated survival signaling pathways, and resistance to radiation-induced apoptosis. The role of SOX9 in maintaining stemness properties strongly suggests that its inhibition could sensitize tumors to radiotherapy by targeting the radioresistant CSC subpopulation.
The mechanisms by which SOX9 inhibition might enhance radiotherapy efficacy include depletion of the cancer stem cell compartment, reversal of therapy-induced stemness, and modulation of the tumor microenvironment. In breast cancer, SOX9 collaborates with Slug (SNAI2) to promote cancer cell proliferation and metastasis [11], processes that may also contribute to radiation resistance. Additionally, SOX9's regulation of epithelial-mesenchymal transition (EMT) [55] represents another potential mechanism, as EMT has been associated with increased radioresistance in various malignancies. Targeting SOX9 may reverse these phenotypic states and restore therapeutic sensitivity.
SOX9 contributes significantly to immunosuppressive tumor microenvironments through multiple mechanisms. In head and neck squamous cell carcinoma (HNSCC), SOX9-enriched tumor cells mediate resistance to combined anti-LAG-3 and anti-PD-1 therapy by regulating the ANXA1-FPR1 axis [27]. Specifically, SOX9 directly regulates annexin A1 (ANXA1) expression, which induces apoptosis of formyl peptide receptor 1 (FPR1)+ neutrophils through mitochondrial fission and inhibited mitophagy. The reduction of FPR1+ neutrophils impairs the infiltration and cytotoxic function of CD8+ T and γδT cells, creating an "immune desert" microenvironment [27]. Similarly, in colorectal cancer, SOX9 expression negatively correlates with infiltration levels of B cells, resting mast cells, resting T cells, monocytes, plasma cells, and eosinophils [4].
Strategic SOX9 inhibition may potentially overcome limitations of conventional immunotherapy, particularly in immunologically "cold" tumors. While immune checkpoint blockers (ICBs) target specific immune pathways, SOX9 inhibition addresses upstream mechanisms that establish broadly immunosuppressive microenvironments. This is evidenced by the enrichment of SOX9+ tumor cells in HNSCC samples resistant to anti-LAG-3 plus anti-PD-1 therapy [27]. The coordinated approach of simultaneously targeting SOX9 while relieving immune suppression may yield superior outcomes compared to sequential or single-modality approaches.
Schematic of SOX9 Inhibition and Therapeutic Combinations: This diagram illustrates the molecular regulation of SOX9, therapeutic combinations with conventional treatments, and resulting biological outcomes.
Table 3: Key Research Reagents for SOX9-Targeted Studies
| Reagent / Tool | Category | Primary Function | Example Applications |
|---|---|---|---|
| THZ2 [56] | Small molecule inhibitor | Covalent CDK7 inhibitor targeting super-enhancers | Reverses TMZ resistance in GBM; suppresses SOX9 expression [56] |
| JQ1 [56] | Small molecule inhibitor | BET bromodomain inhibitor targeting BRD4 | Synergistic antitumor effects with TMZ in GBM [56] |
| Evodiamine [55] | Natural compound inhibitor | Disrupts USP4-mediated SOX9 stabilization | Inhibits colorectal cancer stemness; promotes SOX9 degradation [55] |
| Anti-SOX9 antibodies [54] | Biological reagent | Detection and quantification of SOX9 protein | Immunohistochemistry, Western blotting for SOX9 expression [54] |
| CRISPR/Cas9 SOX9 knockout [54] | Genetic tool | Complete SOX9 gene ablation | Establish causal relationship between SOX9 and chemoresistance [54] |
| siRNA/shSOX9 [55] | Genetic tool | Transient SOX9 knockdown | Functional studies of SOX9 depletion; stemness assays [55] |
| CUT&RUN assay [56] | Epigenetic tool | Mapping transcription factor binding | Identify SOX9 genomic targets; super-enhancer interactions [56] |
| 2-Furancarboxaldehyde, 4-nitro- | 2-Furancarboxaldehyde, 4-nitro-, CAS:57500-49-9, MF:C5H3NO4, MW:141.08 g/mol | Chemical Reagent | Bench Chemicals |
The strategic integration of SOX9 inhibition with conventional chemotherapy and radiotherapy represents a promising frontier in oncology therapeutics, particularly for aggressive, treatment-resistant malignancies. Current evidence demonstrates that SOX9 functions as a critical node regulating cancer stemness, therapeutic resistance, and immune suppression across diverse cancer types. Targeting SOX9 through direct transcriptional inhibition, super-enhancer disruption, or protein stabilization interference can resensitize tumors to conventional treatments and potentially overcome limitations of standalone immunotherapies. While significant progress has been made in elucidating the mechanistic basis for these combinatorial approaches, translation into clinical applications requires further refinement of targeting strategies, patient selection criteria, and optimal sequencing with established treatment modalities. The continued development of sophisticated research tools and experimental models will be essential for advancing this therapeutic paradigm toward meaningful clinical impact.
The combination therapy targeting the immune checkpoint proteins PD-1 and LAG-3 represents a significant advancement in cancer immunotherapy. Despite promising clinical results, a substantial proportion of patients develop resistance to this treatment. Recent research has identified the transcription factor SOX9 (SRY-related HMG-box 9) as a pivotal regulator of resistance mechanisms to anti-PD-1/anti-LAG-3 therapy [27] [3]. This review objectively compares the tumor microenvironment and treatment outcomes between SOX9-high and SOX9-low expressing tumors, providing a comprehensive analysis of supporting experimental data. Framed within the broader thesis of SOX9 inhibition strategies versus conventional immunotherapy, this guide synthesizes current findings for researchers and drug development professionals seeking to overcome immunotherapy resistance.
Single-cell RNA sequencing (scRNA-seq) analyses of resistant head and neck squamous cell carcinoma (HNSCC) mouse models have revealed a novel mechanism by which SOX9+ tumor cells mediate immunotherapy resistance. These cells demonstrate significant enrichment in treatment-resistant samples and directly regulate the expression of annexin A1 (Anxa1) [27].
The established pathway operates as follows:
This mechanism creates an immunosuppressive niche that facilitates tumor escape from immune surveillance despite combined PD-1/LAG-3 blockade.
Figure 1: SOX9-Anxa1-Fpr1 Axis in Immunotherapy Resistance. SOX9 transcriptionally regulates Anxa1, which binds to Fpr1 on neutrophils, ultimately impairing cytotoxic T cell recruitment and function.
Beyond the specific Anxa1-Fpr1 mechanism, SOX9 orchestrates broader immunosuppressive effects within the tumor microenvironment. In lung adenocarcinoma (LUAD) models, SOX9 expression creates an "immune cold" condition characterized by:
These changes establish a tumor microenvironment that is fundamentally resistant to T-cell-mediated killing, which is essential for effective anti-PD-1/anti-LAG-3 therapy.
Table 1: Comparative Analysis of SOX9-High versus SOX9-Low Tumors in Preclinical Models
| Parameter | SOX9-High Tumors | SOX9-Low Tumors | Experimental Model | Citation |
|---|---|---|---|---|
| Response to Anti-PD-1/Anti-LAG-3 | 42.9% resistance rate | 57.1% sensitivity rate | HNSCC mouse model | [27] |
| Tumor Infiltration of CD8+ T cells | Significantly reduced | Normal infiltration | KRAS-driven LUAD model | [21] |
| Overall Survival | Significantly shorter | Significantly longer | KRAS-driven LUAD model | [21] |
| Tumor Grade Distribution | Higher grade (Grade 3) tumors | Predominantly lower grade (Grade 1-2) tumors | GEMM LUAD model | [21] |
| Ki67+ Proliferating Cells | Significantly increased | Reduced percentage | Multiple cancer models | [27] [21] |
Table 2: Immune Microenvironment Composition in SOX9-High Versus SOX9-Low Tumors
| Immune Cell Population | SOX9-High Tumors | SOX9-Low Tumors | Correlation with SOX9 | Cancer Types Observed |
|---|---|---|---|---|
| CD8+ T cells | Decreased infiltration and cytotoxicity | Normal infiltration and function | Negative | LUAD, HNSCC [27] [21] |
| Neutrophils | Reduced Fpr1+ subset, impaired function | Normal accumulation | Negative (Fpr1+ subset) | HNSCC [27] |
| Dendritic Cells | Suppressed infiltration | Normal presence | Negative | LUAD [21] |
| Natural Killer (NK) Cells | Reduced activity and infiltration | Functional anti-tumor activity | Negative | LUAD [21] |
| γδT Cells | Impaired tumor-killing ability | Effective tumor cell killing | Negative | HNSCC [27] |
| Tregs, M2 Macrophages | Context-dependent increase | Normal levels | Positive in some contexts | Multiple cancers [4] |
Experimental Objective: To evaluate resistance mechanisms to anti-LAG-3 and anti-PD-1 combination therapy in head and neck squamous cell carcinoma [27].
Methodology Details:
Experimental Objective: To determine the causal role of SOX9 in lung tumor development and immunotherapy response [21].
Methodology Details:
Figure 2: Experimental Workflow for Investigating SOX9 in Immunotherapy Resistance. Two primary model systems (HNSCC and LUAD) with distinct induction and manipulation methods converge on multi-modal analysis approaches.
Table 3: Key Research Reagents for Investigating SOX9 in Immunotherapy Resistance
| Reagent/Category | Specific Examples | Research Application | Function in Experimental Design |
|---|---|---|---|
| Animal Models | C57BL/6 wild-type mice; KrasLSL-G12D;Sox9flox/flox GEMM | In vivo tumor studies | Provide physiologically relevant systems for studying therapy resistance and SOX9 manipulation [27] [21] |
| Tumor Induction Agents | 4-nitroquinoline 1-oxide (4NQO) | HNSCC model development | Chemical carcinogen that recapitulates human HNSCC pathogenesis [27] |
| Immunotherapy Antibodies | Anti-PD-1, Anti-LAG-3 (relatlimab) | Treatment groups | Immune checkpoint blockade to assess therapeutic efficacy and resistance mechanisms [27] |
| Genetic Manipulation Tools | CRISPR/Cas9 (pSECC system), Cre-LoxP technology | Sox9 gain/loss-of-function studies | Enable precise genetic manipulation of Sox9 in established tumor models [21] |
| Single-Cell Analysis Platforms | scRNA-seq, CopyKAT algorithm | Tumor microenvironment characterization | Identify cell subpopulations and transcriptional programs associated with resistance [27] |
| Cell Tracking Systems | tdTomato reporter | Control and validation | Control for transduction efficiency and track tumor cell populations [21] |
| Cell Culture Models | 3D tumor organoid systems | In vitro mechanistic studies | Provide controlled systems for assessing Sox9 function in tumor growth [21] |
The compiled evidence positions SOX9 as a master regulator of resistance to anti-PD-1/anti-LAG-3 combination therapy through multiple mechanistic pathways. The comparative data clearly demonstrate that SOX9-high tumors exhibit significantly different microenvironments and treatment responses compared to SOX9-low tumors. The emergence of SOX9 as a central node in therapy resistance pathways offers promising directions for diagnostic biomarker development and combination therapeutic strategies. Future research focusing on SOX9 inhibition alongside conventional immunotherapy may potentially overcome resistance mechanisms and improve outcomes for cancer patients.
{ article }
The SOX9-Annexin A1-Fpr1 axis has been identified as a critical mechanism of resistance to combination cancer immunotherapy. Recent research demonstrates that in resistant head and neck squamous cell carcinoma (HNSCC), SOX9+ tumor cells upregulate Annexin A1 (Anxa1), which mediates apoptosis of formyl peptide receptor 1 (Fpr1)+ neutrophils through mitochondrial pathway disruption. This impairs cytotoxic T cell infiltration and creates an immunosuppressive tumor microenvironment. This analysis compares the emerging approach of SOX9 pathway inhibition against conventional immunotherapy, providing experimental data and methodological guidance for researchers investigating this novel resistance pathway.
Combination immunotherapy with anti-PD-1 and anti-LAG-3 antibodies has demonstrated significant efficacy across various cancers, yet a substantial proportion of patients develop resistance through mechanisms that remain incompletely understood [58]. The transcription factor SOX9, known for its roles in development and carcinogenesis, has emerged as a key player in therapy resistance [20] [3]. Recent findings reveal that SOX9 mediates immunosuppression through a novel pathway involving Annexin A1 (Anxa1) and its receptor Fpr1 on neutrophils [58]. This guide provides a comprehensive comparison between conventional immunotherapies and the emerging approach of targeting the SOX9-Anxa1-Fpr1 axis, with detailed experimental data and methodologies to facilitate research in this rapidly advancing field.
The SOX9-Anxa1-Fpr1 axis represents a sophisticated mechanism of immune evasion in treatment-resistant tumors. In resistant HNSCC samples, SOX9+ tumor cells significantly upregulate Anxa1 expression [58]. This protein interacts with Fpr1 receptors on neutrophils, triggering a cascade of intracellular events that promote mitochondrial fission and inhibit mitophagy through downregulation of BCL2/adenovirus E1B interacting protein 3 (Bnip3) [58]. The subsequent induction of neutrophil apoptosis prevents neutrophil accumulation in tumor tissues, which in turn impairs the infiltration and cytotoxic capabilities of CD8+ T and γδT cells within the tumor microenvironment [58].
Table 1: Key Components of the SOX9-Anxa1-Fpr1 Immunosuppressive Axis
| Component | Full Name | Function in the Axis | Expression Pattern |
|---|---|---|---|
| SOX9 | SRY-box transcription factor 9 | Master regulator that directly controls Anxa1 transcription | Upregulated in epithelial tumor cells of resistant cancers |
| Annexin A1 (Anxa1) | Annexin A1 | Effector protein that binds Fpr1 on neutrophils | Highly expressed in SOX9+ resistant tumor cells |
| Fpr1 | Formyl peptide receptor 1 | G-protein coupled receptor on neutrophils that transduces Anxa1 signal | Expressed on tumor-infiltrating neutrophils |
| Bnip3 | BCL2/adenovirus E1B interacting protein 3 | Regulator of mitophagy and mitochondrial function | Downstream target, downregulated upon pathway activation |
The SOX9-Anxa1 interaction represents a crucial switch in immune regulation. In the cancer context, Anxa1 exhibits paradoxical pro-tumoral properties despite its traditional anti-inflammatory functions [59]. This pathway inhibits the accumulation of neutrophils in tumor tissues, subsequently reducing the infiltration and tumor-cell killing ability of cytotoxic CD8+ T cells and γδT cells [58]. Beyond this specific mechanism, SOX9 has broader implications in cancer biology, functioning as either a proto-oncogene or tumor suppressor depending on context [20]. It is significantly upregulated in fifteen cancer types, including GBM, COAD, and LIHC, while being downregulated in only two (SKCM and TGCT) [20].
Visual Abstract 1: SOX9-Anxa1-Fpr1 Signaling Pathway in Immunotherapy Resistance. The diagram illustrates how SOX9 upregulation in tumor cells triggers Anxa1 production, which engages Fpr1 on neutrophils to induce apoptosis through mitochondrial dysfunction, ultimately leading to impaired cytotoxic cell infiltration and therapy resistance.
Targeting the SOX9-Anxa1-Fpr1 axis represents a fundamentally different approach from conventional immunotherapy. While anti-PD-1/LAG-3 therapies aim to directly reinvigorate exhausted T cells, SOX9 pathway inhibition addresses a novel resistance mechanism centered on neutrophil-mediated immunosuppression [58].
Table 2: SOX9 Pathway Inhibition vs. Conventional Immunotherapy Approaches
| Parameter | SOX9 Pathway Inhibition | Anti-PD-1/LAG-3 Therapy | Chemotherapy |
|---|---|---|---|
| Primary Target | SOX9 transcription factor or downstream Anxa1-Fpr1 axis | PD-1/LAG-3 immune checkpoints on T cells | Rapidly dividing cells |
| Mechanism of Action | Prevents neutrophil apoptosis and restores cytotoxic cell infiltration | Blocks inhibitory signals to enhance T cell function | DNA damage or cell cycle disruption |
| Resistance Mechanism | SOX9 overexpression and Anxa1-mediated neutrophil suppression | Upregulation of alternative checkpoints, T cell exhaustion | Drug efflux, DNA repair activation |
| Key Biomarkers | SOX9+ tumor cells, Anxa1 expression, Fpr1+ neutrophils | PD-L1 expression, T cell infiltration | Proliferation markers |
| Therapeutic Context | Emerging target with preclinical validation | FDA-approved for multiple cancers | Standard care for decades |
The comparative efficacy of these approaches reveals complementary strengths. In HNSCC mouse models, anti-LAG-3 plus anti-PD-1 combination therapy initially demonstrated significant efficacy, with 57.1% of animals responding [58]. However, 42.9% developed resistance characterized by significant enrichment of SOX9+ tumor cells [58]. Single-cell RNA sequencing of resistant samples revealed that the proportion of immune cells was dramatically increased in therapy-sensitive groups compared to control or resistant groups, highlighting the significance of tumor-infiltrating immune cells in treatment response [58].
Beyond HNSCC, SOX9 has demonstrated prognostic significance across multiple malignancies. High SOX9 expression correlates with worst overall survival in LGG, CESC, and THYM, suggesting utility as a prognostic marker [20]. In glioblastoma, SOX9 was identified as a diagnostic and prognostic biomarker, particularly in IDH-mutant cases, with expression closely correlated with immune infiltration and checkpoint expression [22].
Research on the SOX9-Anxa1-Fpr1 axis employs sophisticated experimental models that enable mechanistic investigation of this resistance pathway:
1. HNSCC Mouse Model for Therapy Resistance:
2. Single-Cell RNA Sequencing Analysis:
3. Transgenic Mouse Validation:
Visual Abstract 2: Experimental Workflow for Investigating SOX9-Anxa1-Fpr1 Axis. The methodology encompasses animal model development, molecular profiling through single-cell sequencing, and validation using transgenic models to establish mechanistic causality.
Investigating the SOX9-Anxa1-Fpr1 axis requires specialized reagents and models. The following table compiles key research tools derived from published studies:
Table 3: Essential Research Reagents for SOX9-Anxa1-Fpr1 Axis Investigation
| Reagent/Model | Specific Type | Research Application | Key Findings Enabled |
|---|---|---|---|
| 4NQO-induced HNSCC Mouse Model | C57BL/6 wild-type mice | Therapy resistance modeling | Established in vivo correlation between SOX9+ tumors and immunotherapy resistance |
| Anti-LAG-3 + Anti-PD-1 Antibodies | Relatlimab + Nivolumab equivalents | Combination therapy assessment | Demonstrated 42.9% resistance rate in HNSCC with SOX9 enrichment |
| Single-Cell RNA Sequencing | 10X Genomics platform | Tumor microenvironment deconvolution | Identified SOX9+ epithelial subclusters in resistant tumors |
| Transgenic Mouse Models | Various Sox9 and Anxa1 modulators | Mechanistic validation | Confirmed SOX9-mediated Anxa1 transcription and Fpr1+ neutrophil apoptosis |
| Cordycepin | Adenosine analog | SOX9 inhibition studies | Demonstrated dose-dependent SOX9 inhibition in 22RV1, PC3, and H1975 cells |
The identification of the SOX9-Anxa1-Fpr1 axis represents a paradigm shift in understanding immunotherapy resistance. This pathway illuminates a previously underappreciated mechanism where tumor cell-intrinsic factors (SOX9 expression) reshape the immune microenvironment through neutrophil manipulation [58]. The translational potential of targeting this axis is substantial, particularly for patients developing resistance to conventional immunotherapy.
Several targeting strategies emerge from current research. Small molecule inhibitors like cordycepin demonstrate the feasibility of modulating SOX9 activity, showing dose-dependent inhibition of both SOX9 protein and mRNA in prostate and lung cancer cells [20]. Alternative approaches could target downstream effectors, including Anxa1 neutralizing antibodies or Fpr1 antagonists, which have shown promise in modulating immune responses in other contexts [60] [61]. The ANXA1-FPR axis has been investigated as a therapeutic target in transplantation immunology, providing precedent for its targetability [60].
From a diagnostic perspective, SOX9 and associated pathway components offer potential biomarkers for identifying patients at risk for therapy resistance. The correlation between high SOX9 expression and poor prognosis in specific cancer types suggests utility in patient stratification [20] [22]. Future research should focus on developing standardized assays for SOX9 pathway activation and validating these biomarkers in prospective clinical trials.
The SOX9-Anxa1-Fpr1 axis represents a clinically significant resistance mechanism to combination immunotherapy, with robust preclinical validation across multiple models. Targeting this pathway offers a promising complementary approach to conventional immunotherapy, particularly for resistant malignancies. The experimental methodologies and reagents outlined provide a foundation for continued investigation into this emerging therapeutic target. As research advances, targeting the SOX9-Anxa1-Fpr1 axis may unlock new therapeutic opportunities for patients who would otherwise face limited options after immunotherapy failure.
{ /article }
The transcription factor SOX9 (SRY-related HMG-box 9) has emerged as a critical regulator of therapeutic resistance across multiple cancer types. Operating as a master molecular switch, SOX9 drives two interconnected processes that enable cancer cells to survive treatment: epithelial-mesenchymal transition (EMT) and the acquisition of stem-like properties. This review synthesizes current experimental evidence comparing the roles of SOX9 in different cancer contexts, with particular emphasis on its function in promoting drug tolerance through the induction of a plastic, stem-like state. Within the broader thesis of SOX9 inhibition versus conventional immunotherapy outcomes, understanding these mechanisms provides a rational foundation for novel therapeutic combinations aimed at overcoming resistance in recalcitrant malignancies.
SOX9 is a member of the SOX family of transcription factors, characterized by a highly conserved high-mobility group (HMG) DNA-binding domain that recognizes the specific DNA motif (A/TA/TCAAA/TG) [17] [62]. The protein contains three critical functional domains: a dimerization domain (DIM), the HMG domain, and a C-terminal transactivation domain (TAC) [63] [64]. This structural configuration enables SOX9 to perform its diverse functions in development and disease.
Recent research has revealed that SOX9 possesses pioneer factor capability, allowing it to bind cognate motifs in closed chromatin and initiate large-scale transcriptional reprogramming [34]. In skin epidermal stem cells, SOX9 binding to closed chromatin at hair follicle enhancers occurs within one week, with nucleosome displacement and chromatin opening following subsequently [34]. This pioneer activity enables SOX9 to divert embryonic epidermal stem cells (EpdSCs) into becoming hair follicle stem cells, and when reactivated in adult EpdSCs, triggers fate switching toward tumorigenic states [34].
The function of SOX9 in carcinogenesis exhibits significant context dependency, with evidence supporting both tumor-promoting and tumor-suppressing roles depending on the tissue type and molecular environment.
Table 1: Context-Dependent Roles of SOX9 in Different Cancers
| Cancer Type | SOX9 Role | Key Mechanisms | Experimental Evidence |
|---|---|---|---|
| High-Grade Serous Ovarian Cancer | Oncogenic, Drives Chemoresistance | Reprograms transcriptional state to stem-like phenotype; induced by platinum chemotherapy | SOX9 knockout increases platinum sensitivity; single-cell RNA-seq shows upregulation after chemotherapy [17] |
| Glioblastoma | Prognostic Biomarker | Correlates with immune infiltration and checkpoint expression; potential diagnostic marker | High expression associated with better prognosis in lymphoid invasion subgroups; independent prognostic factor for IDH-mutant cases [22] [15] |
| Colorectal Cancer | Tumor Suppressor | Inhibits EMT and stemness; loss promotes invasion and metastasis | Combined Apc and Sox9 inactivation in mice instigates invasive tumors with EMT and SOX2 upregulation [65] |
| Head and Neck Squamous Cell Carcinoma | Mediator of Immunotherapy Resistance | Regulates ANXA1 expression to mediate apoptosis of FPR1+ neutrophils | Sox9+ tumor cells enriched in anti-LAG-3+anti-PD-1 resistant samples; directly regulates Anxa1 expression [27] |
| Skin Cancer | Pioneer Factor in Fate Switching | Reprograms epidermal stem cells to hair follicle stem cells; sustained expression leads to BCC | SOX9 binding to closed chromatin precedes chromatin opening and nucleosome displacement [34] |
In high-grade serous ovarian cancer (HGSOC), SOX9 functions unequivocally as an oncogene and key mediator of platinum resistance. Treatment of HGSOC cell lines (OVCAR4, Kuramochi, and COV362) with carboplatin induced robust SOX9 upregulation at both RNA and protein levels within 72 hours [17]. Clinical validation using longitudinal single-cell RNA-Seq data from 11 HGSOC patients demonstrated that SOX9 expression significantly increased in post-neoadjuvant chemotherapy (NACT) tissues compared to treatment-naive samples, with 8 of 11 patients showing elevated SOX9 after chemotherapy [17]. Functional studies confirmed that SOX9 ablation increased platinum sensitivity, while its overexpression induced a stem-like subpopulation and significant chemoresistance in vivo [17].
SOX9 promotes epithelial-mesenchymal transition through multiple interconnected signaling pathways. In colorectal cancer models, Sox9 inactivation promoted EMT pathway activation and SOX2 stem cell factor upregulation [65]. The loss of SOX9 in Apc-deficient mouse colon tumors instigated more invasive tumors with clear EMT characteristics, while human CRCs with low SOX9 expression showed prominent EMT pathway gene expression changes [65]. This positions SOX9 as a critical regulator of the epithelial-mesenchymal balance in specific tissue contexts.
Beyond EMT, SOX9 directly regulates stemness properties that confer drug tolerance. In HGSOC, SOX9 increases transcriptional divergence, a metric measuring overall transcriptional malleability defined as the sum of expression of the top 50% of detected genes divided by the sum of expression of the bottom 50% (P50/P50) [17]. This transcriptional state represents a cell's ability to respond effectively to external stressors like chemotherapy and is amplified in stem cells and cancer stem cells (CSCs) [17]. A rare cluster of SOX9-expressing cells identified in primary HGSOC tumors was highly enriched for CSCs and chemoresistance-associated stress gene modules [17].
Table 2: Experimental Evidence for SOX9 in Stemness and Drug Resistance
| Experimental System | Key Finding | Methodology | Reference |
|---|---|---|---|
| HGSOC cell lines and patient-derived xenografts | SOX9 induces stem-like transcriptional state and platinum resistance | Multiomics, tumor microarrays, epigenetic modulation, single-cell analysis | [17] |
| Mouse epidermal stem cell model | SOX9 acts as pioneer factor to switch stem cell fates | Genetic lineage tracing, CUT&RUN, ATAC-seq, chromatin dynamics | [34] |
| Colorectal cancer mouse models | SOX9 loss promotes stemness via SOX2 upregulation | Conditional knockout mice, IHC, transcriptional profiling, survival analysis | [65] |
| Head and neck cancer mouse model | Sox9+ tumor cells mediate immunotherapy resistance | Single-cell RNA sequencing, transgenic models, immune cell profiling | [27] |
In ovarian cancer, SOX9 emerges as a critical early responder to platinum agents, driving a transcriptional program that fosters tolerance. Mechanistically, epigenetic upregulation of SOX9 was sufficient to induce chemoresistance in multiple HGSOC lines, while its ablation sensitized cells to platinum treatment [17]. Single-cell analysis demonstrated that chemotherapy treatment results in rapid population-level induction of SOX9 that enriches for a stem-like transcriptional state [17]. This positions SOX9 as a key regulator of early steps of transcriptional reprogramming that lead to chemoresistance through a CSC-like state in HGSOC.
In head and neck squamous cell carcinoma (HNSCC), SOX9 mediates resistance to combination immune checkpoint blockade (anti-LAG-3 plus anti-PD-1) through a novel mechanism involving the tumor microenvironment [27]. scRNA-seq of resistant tumors revealed significant enrichment of Sox9+ tumor cells, which directly regulate the expression of annexin A1 (Anxa1) [27]. This SOX9-Anxa1 axis mediates apoptosis of formyl peptide receptor 1 (Fpr1)+ neutrophils by promoting mitochondrial fission and inhibiting mitophagy through downregulation of Bnip3 expression [27]. The consequent reduction of Fpr1+ neutrophils impairs the infiltration and tumor cell-killing ability of cytotoxic Cd8 T and γδT cells within the tumor microenvironment, ultimately driving resistance [27].
Research into SOX9 function employs sophisticated genetic and molecular techniques. For lineage tracing and fate mapping in skin models, researchers engineered mice harboring a MYC-epitope-tagged Sox9 transgene controlled by a tetracycline-responsive enhancer and minimal promoter (TRE-Sox9), which were bred to lines expressing the tetracycline-inducible transcriptional activator (rtTA) driven by an epidermal (Krt14) promoter [34]. This system allowed precise temporal control over SOX9 re-activation in adult epidermal stem cells to monitor reprogramming events [34].
For chromatin dynamics studies, the CUT&RUN (cleavage under targets and release using nuclease) sequencing method was employed to temporally assay SOX9 binding to chromatin, while ATAC-seq (assay for transposase-accessible chromatin with high-throughput sequencing) interrogated chromatin accessibility during reprogramming [34]. These techniques revealed that SOX9 binding to chromatin preceded increased accessibility, demonstrating its pioneer factor capability [34].
Table 3: Key Research Reagents for SOX9 Investigation
| Reagent/Resource | Function/Application | Experimental Context |
|---|---|---|
| CDX2P-CreERT2 transgenic mice | Tamoxifen-inducible Cre recombinase for colon-specific gene targeting | Colorectal cancer models for Apc and Sox9 inactivation [65] |
| Krt14-rtTA;TRE-Sox9 mice | Doxycycline-inducible SOX9 expression in epidermal stem cells | Skin reprogramming and BCC model [34] |
| CRISPR/Cas9 with SOX9-targeting sgRNA | SOX9 knockout for functional validation | HGSOC lines for platinum sensitivity assays [17] |
| Single-cell RNA sequencing | Transcriptomic profiling at single-cell resolution | Identification of Sox9+ subpopulations in resistant tumors [17] [27] |
| CUT&RUN sequencing | Mapping transcription factor binding sites | SOX9 chromatin occupancy studies [34] |
| ATAC-seq | Assessing chromatin accessibility | Nucleosome displacement and chromatin remodeling by SOX9 [34] |
The diagrams below visualize key signaling pathways and experimental workflows described in the research.
The accumulated evidence positions SOX9 as a promising therapeutic target to overcome drug tolerance, particularly in combination with conventional therapies. Several strategic approaches emerge for targeting SOX9:
Direct SOX9 targeting remains challenging due to the difficulty of inhibiting transcription factors with small molecules, though emerging technologies like proteolysis-targeting chimeras (PROTACs) may offer solutions. Indirect approaches include targeting SOX9-upregulated surface markers on CSCs, disrupting SOX9-cofactor interactions, or modulating upstream regulators of SOX9 expression [63].
In HNSCC, where SOX9 mediates resistance to anti-LAG-3+anti-PD-1 therapy, targeting the SOX9-Anxa1-Fpr1-neutrophil axis may restore therapeutic sensitivity [27]. In ovarian cancer, SOX9 inhibition during platinum-based chemotherapy could prevent the emergence of drug-tolerant persister cells [17]. For colorectal cancers with SOX9 loss, strategies to restore SOX9 function might suppress EMT and metastasis [65].
SOX9 stands as a critical node in the network of therapy resistance, integrating signals from the microenvironment to drive EMT and stemness programs that enable cancer cell survival under therapeutic pressure. Its context-dependent functionsâas either oncogene or tumor suppressorâhighlight the necessity for precise patient stratification in therapeutic targeting. Future research directions should focus on elucidating the determinants of SOX9's dualistic nature, developing clinically viable SOX9 inhibitors, and designing rational combination therapies that simultaneously target SOX9-mediated resistance pathways alongside standard-of-care treatments. Within the broader thesis of SOX9 inhibition versus conventional immunotherapy, the evidence suggests that SOX9-targeted approaches may prove most effective as adjuvants to existing modalities, potentially overcoming the drug tolerance that limits current cancer therapeutics.
The transcription factor SOX9 plays a pivotal role in shaping the tumor immune microenvironment, particularly through its regulation of immune checkpoint pathways and promotion of immunologically "cold" tumors. This review synthesizes evidence from recent studies demonstrating how SOX9 orchestrates multiple mechanisms to create immune-suppressive landscapes that facilitate tumor immune evasion. By examining comparative data across cancer types and experimental models, we establish a framework for understanding SOX9 inhibition as a promising strategy to overcome limitations of conventional immunotherapies. The compiled findings reveal consistent patterns of SOX9-mediated immune exclusion through direct regulation of checkpoint molecules, suppression of cytotoxic immune cell infiltration, and alteration of the tumor extracellular matrix. This analysis provides a foundation for developing novel therapeutic approaches that target SOX9 to convert immune-desert tumors into immune-inflamed microenvironments responsive to checkpoint inhibition.
The SRY-box transcription factor 9 (SOX9) has emerged as a critical regulator of tumor immunobiology, operating at the intersection of cancer cell intrinsic signaling and extrinsic immune modulation. While initially characterized for its roles in development and differentiation, SOX9 is frequently overexpressed in diverse malignancies including lung, breast, colorectal, and head and neck cancers, where its expression often correlates with poor prognosis and therapy resistance [4] [20] [3]. Beyond its established functions in tumor proliferation and metastasis, accumulating evidence positions SOX9 as a central orchestrator of the tumor immune microenvironment (TIME), particularly through the formation of "immune desert" phenotypes characterized by absent or excluded cytotoxic immune cells [57] [66].
The immune desert phenotype represents one of three broad classifications of tumor immune landscapes, alongside immune-inflamed and immune-excluded patterns. While immune-inflamed tumors show abundant T cell infiltration both in tumor nests and stroma, and immune-excluded tumors display T cells restricted to peritumoral regions, immune-desert tumors exhibit a profound absence of T cells throughout the tumor core and periphery [66]. This classification has profound therapeutic implications, as immune-desert tumors typically demonstrate minimal response to immune checkpoint inhibitors (ICIs) due to the lack of pre-existing anti-tumor immunity [37] [66].
This review systematically analyzes the mechanisms through which SOX9 regulates immune checkpoint pathways and drives the formation of immune-desert tumors. By integrating findings from preclinical models and human tumor analyses, we provide a comparative assessment of SOX9-targeting strategies versus conventional immunotherapy approaches, offering insights for researchers and drug development professionals seeking to overcome immunotherapy resistance.
Comprehensive analyses of SOX9 expression across human cancers reveal distinctive patterns with significant implications for tumor immunity. Bioinformatic investigations using data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases demonstrate that SOX9 expression is significantly elevated in 15 of 33 cancer types compared to matched normal tissues, including cervical cancer (CESC), colorectal adenocarcinoma (COAD), glioblastoma (GBM), lung squamous cell carcinoma (LUSC), pancreatic adenocarcinoma (PAAD), and others [20]. Notably, only two cancer typesâskin cutaneous melanoma (SKCM) and testicular germ cell tumors (TGCT)âshow significantly decreased SOX9 expression, highlighting its generally oncogenic character across most malignancies [20].
The prognostic significance of SOX9 varies by cancer type, with high expression correlating with worse overall survival in low-grade glioma (LGG), CESC, and thymoma (THYM), but surprisingly with better prognosis in adrenocortical carcinoma (ACC) [20]. In glioma, SOX9 has been identified as both a diagnostic and prognostic biomarker, particularly in isocitrate dehydrogenase (IDH)-mutant cases where its expression closely correlates with immune infiltration patterns and checkpoint molecule expression [22].
The relationship between SOX9 expression and immune cell infiltration follows distinct patterns across cancer types, as summarized in Table 1. Integrated analyses of whole exome and RNA sequencing data from TCGA reveal that SOX9 expression negatively correlates with infiltration of B cells, resting mast cells, resting T cells, monocytes, plasma cells, and eosinophils in colorectal cancer [4]. Conversely, SOX9 shows positive correlation with neutrophils, macrophages, activated mast cells, and naive/activated T cells [4].
Table 1: Correlation Between SOX9 Expression and Immune Cell Infiltration Across Cancer Types
| Cancer Type | Immune Cells Positively Correlated with SOX9 | Immune Cells Negatively Correlated with SOX9 | Clinical Implications |
|---|---|---|---|
| Colorectal Cancer | Neutrophils, macrophages, activated mast cells, naive/activated T cells | B cells, resting mast cells, resting T cells, monocytes, plasma cells, eosinophils | Associated with immune exclusion and poor prognosis [4] |
| Lung Adenocarcinoma | M2 macrophages, Tregs | CD8+ T cells, natural killer (NK) cells, dendritic cells, M1 macrophages | Creates "immune cold" microenvironment resistant to ICIs [57] [21] |
| Prostate Cancer | Tregs, M2 macrophages, anergic neutrophils | CD8+ CXCR6+ T cells, activated neutrophils | Androgen deprivation therapy enriches SOX9+ club cells [4] |
| Glioblastoma | Variable by subtype | Variable by subtype | High SOX9 associated with better prognosis in lymphoid invasion subgroups [22] |
| Head and Neck SCC | Sox9+ tumor cells, Fpr1+ neutrophils (via ANXA1) | Cytotoxic CD8+ T cells, γδT cells | Mediates resistance to anti-LAG-3 + anti-PD-1 therapy [27] |
In lung adenocarcinoma (LUAD), SOX9 significantly suppresses infiltration of multiple anti-tumor immune populations. Analysis of KRAS-driven murine LUAD models demonstrates that SOX9 negatively impacts CD8+ T cells, natural killer (NK) cells, and dendritic cells, while concurrently elevating collagen-related gene expression and increasing collagen fiber deposition [21]. This combination of immune suppression and extracellular matrix remodeling creates a physical and biological barrier to immune cell infiltration, characteristic of immune-desert or immune-excluded phenotypes.
SOX9 influences the expression of several immune checkpoint molecules that modulate T cell function and anti-tumor immunity. In thymoma, SOX9 expression negatively correlates with genes associated with PD-L1 expression and T-cell receptor signaling pathways, suggesting its involvement in checkpoint regulation [20]. Similarly, in breast cancer, SOX9 facilitates immune escape by triggering tumorigenesis through mechanisms that likely involve checkpoint modulation [23].
The interaction between SOX9 and checkpoint pathways extends beyond PD-1/PD-L1. Recent research in head and neck squamous cell carcinoma (HNSCC) reveals that SOX9+ tumor cells mediate resistance to combination therapy targeting both PD-1 and LAG-3, another critical immune checkpoint [27]. Single-cell RNA sequencing of treatment-resistant HNSCC samples shows significant enrichment of SOX9+ tumor cells that directly regulate annexin A1 (ANXA1) expression, which in turn mediates apoptosis of formyl peptide receptor 1 (FPR1)+ neutrophils through the ANXA1-FPR1 axis [27]. This pathway ultimately impairs infiltration and cytotoxic function of CD8+ T and γδT cells within the tumor microenvironment [27].
Figure 1: SOX9-ANXA1-FPR1 Axis in Immunotherapy Resistance. SOX9 upregulates ANXA1 expression, which activates FPR1 on neutrophils, promoting mitochondrial fission and inhibiting mitophagy through BNIP3 downregulation, ultimately reducing neutrophil accumulation and impairing cytotoxic cell function.
Beyond direct checkpoint regulation, SOX9 employs multiple mechanisms to suppress immune cell infiltration and function within the tumor microenvironment. In KRAS-mutant lung cancer, SOX9 overexpression creates an "immune cold" condition characterized by markedly reduced T cell infiltration [57]. This effect appears to be SOX9-specific, as knockout of Sox9 delays tumor formation while overexpression accelerates it, with profound effects on immune cell recruitment [57].
The mechanisms underlying SOX9-mediated immune exclusion include:
Extracellular Matrix Remodeling: SOX9 significantly elevates collagen-related gene expression and increases collagen fiber deposition in lung adenocarcinoma, creating a physical barrier to immune cell infiltration [21]. This matrix remodeling contributes to the immune-excluded phenotype, where T cells are present in stromal regions but unable to penetrate tumor nests.
Myeloid Cell Modulation: SOX9 suppresses tumor-associated dendritic cells, which are essential for antigen presentation and T cell priming [21]. By inhibiting dendritic cell function, SOX9 disrupts the cancer-immunity cycle at its initial stages, preventing the generation of effective anti-tumor T cell responses.
Cytokine and Chemokine Regulation: SOX9 influences the expression of various chemokines and cytokines that coordinate immune cell trafficking and function. Though the specific factors vary by cancer type, the net effect is reduced attraction and activation of cytotoxic lymphocytes while potentially promoting immunosuppressive cell populations.
SOX9 further shapes the immunosuppressive landscape by promoting the expansion and activity of regulatory immune cells. Across multiple cancer types, including prostate cancer and lung adenocarcinoma, SOX9 expression correlates with increased infiltration of regulatory T cells (Tregs) and M2 macrophages [4] [37]. These cells actively suppress effector T cell function through multiple mechanisms including cytokine secretion (IL-10, TGF-β), metabolic disruption (adenosine, IDO), and direct inhibition.
In liver cancer, the related SOX family member SOX18 promotes accumulation of Tregs and immunosuppressive tumor-associated macrophages (TAMs) by transactivating PD-L1 and CXCL12 [37]. While direct evidence for SOX9 in this specific mechanism requires further investigation, the conservation of function among SOX proteins suggests similar activities for SOX9 in appropriate cellular contexts.
Various experimental models have been employed to elucidate SOX9's role in immune regulation, each offering distinct advantages for mechanistic studies. Table 2 summarizes key methodologies and findings from recent investigations.
Table 2: Experimental Models for Studying SOX9 in Tumor Immunity
| Experimental Model | Key Methodologies | Major Findings | Reference |
|---|---|---|---|
| KrasLSL-G12D; Sox9flox/flox GEMM | CRISPR/Cas9 and Cre-LoxP gene knockout; flow cytometry; IHC; organoid culture | Sox9 loss reduced tumor development, burden, and progression; increased survival; suppressed immune cell infiltration | [21] |
| 4NQO-induced HNSCC mouse model | Single-cell RNA sequencing; immunohistochemistry; magnetic resonance imaging; transgenic models | Sox9+ tumor cells enriched in anti-LAG-3+anti-PD-1 resistant tumors; ANXA1-FPR1 axis mediates neutrophil apoptosis | [27] |
| KRAS-mutant lung cancer model | Sox9 knockout and overexpression; tumor grafting in immunocompetent vs. immunocompromised mice | Sox9 creates "immune cold" tumors; regulates collagen deposition and immune cell infiltration | [57] |
| Pan-cancer bioinformatic analysis | TCGA and GTEx data analysis; correlation with immunomodulators and immune infiltration | SOX9 upregulated in 15/33 cancer types; correlates with distinct immune cell populations | [20] |
| Prostate cancer xenograft | Cordycepin treatment; Western blot; RNA analysis; cell proliferation assays | Cordycepin inhibits SOX9 expression in dose-dependent manner; potential therapeutic approach | [20] |
Investigating SOX9-mediated immune regulation requires specialized reagents and methodologies. Table 3 outlines key research tools for studying SOX9 in the tumor immune microenvironment.
Table 3: Essential Research Reagents for SOX9-Immunity Studies
| Reagent/Resource | Function/Application | Example Use | Source |
|---|---|---|---|
| KrasLSL-G12D; Sox9flox/flox mice | Genetically engineered mouse model for conditional Sox9 knockout in KRAS-driven tumors | Studying Sox9 loss effects on tumor development and immune infiltration in LUAD | [21] |
| Anti-LAG-3 + Anti-PD-1 antibodies | Immune checkpoint blockade combination therapy | Evaluating therapy resistance mechanisms in HNSCC | [27] |
| Single-cell RNA sequencing | High-resolution characterization of tumor immune microenvironment | Identifying Sox9+ tumor subpopulations in treatment-resistant samples | [27] |
| Cordycepin | Natural compound that inhibits SOX9 expression | Testing SOX9 targeting as therapeutic strategy in prostate cancer | [20] |
| CopyKAT algorithm | Computational tool for identifying malignant cells from scRNA-seq data | Distinguishing malignant from non-malignant epithelial cells | [27] |
| TCGA and GTEx databases | Human tumor and normal tissue gene expression data | Pan-cancer analysis of SOX9 expression and immune correlations | [20] [22] |
Conventional immunotherapies, particularly immune checkpoint inhibitors (ICIs), have revolutionized cancer treatment but face significant limitations in immune-desert tumors. The response rates to ICIs monotherapy range from 10-58% across cancer types, with minimal activity in immune-excluded and immune-desert microenvironments [66]. The IMPassion130 trial revealed declining PD-L1 expression following the progression from immune-inflamed to immune-excluded and immune-desert phenotypes, explaining the reduced efficacy of ICIs in these contexts [66].
In HNSCC, combination therapy targeting both PD-1 and LAG-3 demonstrates improved outcomes compared to monotherapy, but resistance develops in a substantial proportion of cases [27]. Single-cell RNA sequencing of resistant tumors reveals significant enrichment of SOX9+ tumor cells, highlighting SOX9 as a key mediator of resistance to even combination checkpoint blockade [27].
Direct and indirect targeting of SOX9 presents a promising alternative or complementary approach to conventional immunotherapy. Several lines of evidence support this strategy:
SOX9 Knockout Models: In Kras-driven lung adenocarcinoma, conditional knockout of Sox9 significantly reduces tumor development, burden, and progression while improving overall survival [21]. The antitumor effect of Sox9 loss is significantly attenuated in immunocompromised mice compared to syngeneic hosts, indicating that functional immune system is required for maximal therapeutic benefit [21].
Small Molecule Inhibition: Cordycepin, an adenosine analog, inhibits both protein and mRNA expression of SOX9 in a dose-dependent manner in prostate cancer cells (22RV1, PC3) and lung cancer cells (H1975) [20]. This inhibition correlates with reduced cancer cell proliferation, suggesting SOX9 targeting as a viable therapeutic strategy.
Combination Approaches: The integral role of SOX9 in multiple resistance mechanisms suggests that its inhibition could synergize with existing immunotherapies. By disrupting the SOX9-mediated immunosuppressive network, tumors may be converted from immune-desert to immune-inflamed phenotypes, thereby sensitizing them to checkpoint inhibition.
Figure 2: SOX9-Targeted Therapy Reverses Immune Desert Phenotype. SOX9 inhibition reverses multiple immunosuppressive mechanisms, potentially converting immune-desert tumors into immune-inflamed microenvironments sensitive to immunotherapy.
The accumulating evidence firmly establishes SOX9 as a critical regulator of immune checkpoint pathways and architect of the immune-desert tumor microenvironment. Through direct regulation of checkpoint molecules, suppression of cytotoxic immune cell infiltration, induction of immunosuppressive populations, and remodeling of the extracellular matrix, SOX9 creates a multifaceted barrier to effective anti-tumor immunity.
The comparative analysis presented herein suggests that SOX9 inhibition represents a promising strategy to overcome limitations of conventional immunotherapies, particularly in immune-desert and immune-excluded tumors. While several technical challenges remainâincluding the difficulty of directly targeting transcription factors and the context-dependent functions of SOX9 across cancer typesâemerging approaches such as cordycepin-mediated SOX9 suppression and combination strategies with existing immunotherapies offer promising avenues for clinical translation.
Future research directions should include: (1) development of more specific and potent SOX9 inhibitors; (2) comprehensive characterization of SOX9's immunomodulatory functions across cancer types; (3) identification of biomarkers predicting response to SOX9-targeted therapies; and (4) optimization of combination regimens with immune checkpoint blockers. By addressing these priorities, researchers and drug development professionals can harness the potential of SOX9 targeting to expand the benefits of cancer immunotherapy to currently resistant populations.
Biomarker-Driven Patient Stratification for SOX9-Targeted Interventions
The transcription factor SOX9 has emerged as a critical biomarker in oncology, with its expression levels providing significant prognostic value across various cancer types. Research demonstrates that SOX9 is frequently dysregulated in tumors and is intricately involved in key cancer hallmarks, including therapy resistance, immune evasion, and stemness. The table below summarizes the prognostic significance of SOX9 expression in selected cancers, underscoring its utility for patient stratification.
Table 1: Prognostic Value of SOX9 Expression in Pan-Cancer Analyses
| Cancer Type | SOX9 Expression vs. Normal | Correlation with Patient Survival | Clinical Implications |
|---|---|---|---|
| Glioblastoma (GBM) | Significantly upregulated [22] [20] | Better prognosis in specific subgroups (e.g., lymphoid invasion); independent prognostic factor for IDH-mutant [22] | Diagnostic and prognostic biomarker; potential therapeutic target [22] |
| Low-Grade Glioma (LGG) | Significantly upregulated [20] | Shorter Overall Survival [20] | Prognostic biomarker indicating high risk [20] |
| Lung Adenocarcinoma (LUAD) | Upregulated in various cancers [22] | Significant correlation with poorer overall survival [22] | Biomarker for tumor grading and prognosis [22] |
| Ovarian Cancer | Upregulated in chemoresistant cells and patient samples [54] | Associated with chemoresistance and poor outcomes [54] | Biomarker for chemotherapy resistance; potential therapeutic target [54] |
| Head and Neck Squamous Cell Carcinoma (HNSCC) | Enriched in therapy-resistant tumors [27] | Mediates resistance to anti-LAG-3 + anti-PD-1 therapy [27] | Predictor of non-response to combination immunotherapy; resistance biomarker [27] |
| Skin Cutaneous Melanoma (SKCM) | Significantly downregulated [20] | Tumor suppressor role; inhibits tumorigenicity [20] | Context-dependent role, highlighting need for precise stratification [20] |
Emerging evidence positions SOX9 inhibition as a promising strategy to overcome the limitations of conventional therapies, particularly chemotherapy and immunotherapy. The following table compares therapeutic outcomes, highlighting the potential of targeting SOX9.
Table 2: Comparison of Therapeutic Outcomes: Conventional Therapy vs. SOX9-Targeted Approaches
| Therapy Modality | Key Mechanism | Limitations / Resistance Mechanisms | Potential of SOX9-Targeting |
|---|---|---|---|
| Chemotherapy (e.g., in Ovarian Cancer) | Cytotoxic cell death | SOX9 upregulation reprograms cancer cells into stem-like, chemoresistant cells [54] | SOX9 knockdown can reverse resistance and suppress tumor growth [3] |
| Anti-PD-1 + Anti-LAG-3 Immunotherapy (e.g., in HNSCC) | Reverses T-cell exhaustion | SOX9+ tumor cells mediate resistance via ANXA1-FPR1 axis, impairing neutrophil and cytotoxic T-cell function [27] | Targeting the SOX9/ANXA1 axis may restore therapeutic sensitivity and immune cell killing [27] |
| Broad-Spectrum Targeted Therapy | Inhibits specific oncogenic pathways | SOX9 activation induces resistance in various cancers (e.g., NSCLC, liver cancer) [3] | SOX9 inhibition can re-sensitize tumors to agents like Sorafenib and EGFR-TKIs [3] |
The development of SOX9-targeted interventions relies on robust experimental models to validate its role and therapeutic potential. Key methodologies are outlined below.
Table 3: Essential Experimental Workflows for SOX9 Research
| Experimental Goal | Standard Protocol / Model | Key Outcome Measurements |
|---|---|---|
| Establishing SOX9's Role in Chemoresistance | In vitro: Treat ovarian cancer cell lines with chemotherapeutics. In vivo: Use patient-derived xenografts (PDXs) [54]. | ⢠SOX9 protein/mRNA expression (Western Blot, qRT-PCR) [54] ⢠Tumor sphere formation assay ⢠Apoptosis rate (e.g., cleaved Caspase-3) [27] ⢠In vivo tumor volume and survival |
| Modeling Immunotherapy Resistance | ⢠Induce HNSCC in C57BL/6 mice with 4-nitroquinoline 1-oxide (4NQO) [27]. ⢠Treat with anti-PD-1 + anti-LAG-3 antibodies. ⢠Stratify into resistant/sensitive groups per RECIST criteria [27]. | ⢠Single-cell RNA sequencing (scRNA-seq) of tumor microenvironment [27] ⢠Immune cell infiltration (flow cytometry, IHC) ⢠Neutrophil apoptosis assays ⢠Mitochondrial fission and mitophagy markers |
| Pan-Cancer Biomarker Analysis | ⢠Bioinformatics analysis of TCGA, GTEx, and HPA databases [22] [20]. ⢠Correlation with clinical data (overall survival, disease-free survival). | ⢠Differential gene expression analysis [22] ⢠Immune cell infiltration estimation (e.g., ssGSEA) [22] ⢠Kaplan-Meier survival curves and COX regression analysis [22] [20] |
| Functional Genetic Perturbation | ⢠CRISPR/Cas9-mediated SOX9 knockout or activation in cancer cell lines [54]. ⢠RNAi-mediated SOX9 knockdown [3]. | ⢠Transcriptome analysis (RNA-seq) [54] ⢠Cell proliferation, invasion, migration assays ⢠Chemosensitivity and drug response tests |
Successful investigation into SOX9 requires a suite of reliable research reagents and tools. The following table details essential materials for this field.
Table 4: Key Research Reagents and Resources for SOX9 Investigations
| Reagent / Resource | Function and Application | Example Use Case |
|---|---|---|
| CRISPR/Cas9 System | For precise gene editing to knockout or activate the SOX9 gene in cell lines. | Functionally validating SOX9's role in chemoresistance in ovarian cancer models [54]. |
| scRNA-seq Platform | To profile transcriptomes of individual cells within a complex tissue, revealing heterogeneity. | Identifying SOX9+ tumor cell subpopulations and their interactions in the tumor immune microenvironment [27]. |
| SOX9 Antibodies (for IHC, WB) | For detecting and quantifying SOX9 protein expression and localization in tissue samples and cell lysates. | Assessing SOX9 upregulation in patient tumor samples pre- and post-chemotherapy [54]. |
| TCGA/GTEx Databases | Publicly available repositories of genomic, transcriptomic, and clinical data from thousands of tumor and normal samples. | Performing pan-cancer analysis of SOX9 expression and its correlation with survival [22] [20]. |
| Cordycepin (CD) | A small-molecule adenosine analog that inhibits SOX9 expression. | Experimentally downregulating SOX9 to study its functional impact and as a potential therapeutic agent [20]. |
| LASSO-COX Regression | A statistical and machine learning method for selecting the most relevant prognostic features from high-dimensional data. | Constructing a robust prognostic model and nomogram incorporating SOX9 and other variables [22]. |
SOX9 drives treatment failure through multiple, context-dependent molecular pathways. The diagrams below illustrate key mechanisms of resistance to chemotherapy and immunotherapy.
Diagram 1: SOX9 Drives Chemoresistance. Chemotherapy stress induces SOX9 upregulation, which acts as a master regulator to reprogram cancer cells into a stem-like state, leading to therapy resistance [54] [3].
Diagram 2: SOX9 Mediates Immunotherapy Resistance. SOX9+ tumor cells transcriptionally upregulate ANXA1, which binds to FPR1 on neutrophils. This interaction induces neutrophil apoptosis and inhibits mitophagy, ultimately impairing cytotoxic T-cell function and driving resistance to combination immunotherapy [27].
The strategic inhibition of SOX9 presents a promising avenue for overcoming resistance to conventional chemotherapy and modern immunotherapies. The evidence confirms that biomarker-driven patient stratificationâbased on SOX9 expression levels, IDH mutation status, and specific cellular contextsâis not merely beneficial but essential for the success of future clinical trials. Future research must focus on developing direct and potent SOX9 inhibitors, validating non-invasive methods to detect SOX9 levels, and designing innovative clinical trials that incorporate these stratification principles from the outset. By integrating robust biomarker signatures, the field can unlock the potential of SOX9-targeted interventions and provide new hope for patients with resistant cancers.
The transcription factor SOX9 (SRY-box 9) is increasingly recognized as a critical regulator of therapeutic resistance across diverse cancer types. Preclinical investigations consistently demonstrate that SOX9 expression drives resistance to conventional chemotherapy, targeted therapy, and emerging immunotherapies through multiple molecular mechanisms. This resistance is mediated through SOX9's roles in promoting a stem-like transcriptional state, enhancing DNA damage response, regulating immune cell function within the tumor microenvironment, and controlling apoptotic pathways. Research across various solid tumors, including ovarian cancer, chondrosarcoma, head and neck squamous cell carcinoma, and glioblastoma, provides compelling evidence that genetic ablation of SOX9 significantly sensitizes tumor cells to conventional therapeutic agents. This review synthesizes experimental data from these preclinical models to objectively compare the therapeutic sensitization effects achieved through SOX9 inhibition, providing a foundation for developing novel combination treatment strategies.
In high-grade serous ovarian cancer (HGSOC), SOX9 emerges as a key chemotherapy-induced driver of chemoresistance. Research demonstrates that SOX9 is epigenetically upregulated following platinum-based chemotherapy, sufficient to induce significant resistance both in vitro and in vivo [17]. Mechanistically, SOX9 increases transcriptional divergence, reprogramming naive cells into a stem-like state characterized by enhanced survival capacity [17]. This SOX9-enriched population shows remarkable enrichment for cancer stem cell (CSC) markers and chemoresistance-associated stress gene modules [17].
Experimental Evidence: CRISPR/Cas9-mediated SOX9 knockout in multiple HGSOC cell lines (OVCAR4, Kuramochi, COV362) significantly increased sensitivity to carboplatin treatment, as quantified by colony formation assays (2-tailed Student's t-test, P = 0.0025) [17]. Single-cell RNA sequencing analysis of patient tumors before and after neoadjuvant chemotherapy confirmed significant SOX9 upregulation post-treatment (Wilcoxon's P < 2.2e-16), validating the clinical relevance of these findings [17].
Table 1: SOX9 Knockout Effects on Chemotherapy Response in Preclinical Models
| Cancer Type | Therapeutic Agent | Experimental Model | Key Findings | Molecular Mechanisms |
|---|---|---|---|---|
| High-Grade Serous Ovarian Cancer | Carboplatin | CRISPR/Cas9 SOX9 knockout in OVCAR4, Kuramochi, COV362 lines | Significant increased sensitivity (P = 0.0025); reduced colony formation | Reduced stem-like transcriptional state; decreased transcriptional divergence |
| Chondrosarcoma | Doxorubicin | CRISPR/Cas9 SOX9 knockout in HTB94 cell line | Increased drug impact; reduced proliferation and clonogenicity | Impaired MMP13 activation; altered BCL2 and survivin expression |
| Chondrosarcoma | Oncolytic Virus | CRISPR/Cas9 SOX9 knockout in HTB94 cell line | Reduced treatment sensitivity | Not fully elucidated; potentially related to altered viral entry/replication |
Chondrosarcoma presents a unique context for SOX9 function, as it is the master transcription factor for chondrogenesis. SOX9 expression is significantly increased in grade I-III chondrosarcoma compared to normal cartilage, though this pattern changes in dedifferentiated chondrosarcoma (DDCS), where SOX9 is almost completely absent in dedifferentiated compartments [67].
Experimental Evidence: CRISPR/Cas9-mediated SOX9 knockout in the human chondrosarcoma cell line HTB94 resulted in substantially altered response to conventional therapies. SOX9-depleted cells demonstrated increased sensitivity to doxorubicin but surprisingly showed reduced sensitivity to oncolytic virus treatment [67]. This bidirectional effect highlights the context-dependent nature of SOX9-mediated resistance. The knockout cells exhibited reduced proliferation, clonogenicity, and migration, alongside increased adhesion, apoptosis, and polyploidy formation [67]. Molecular analysis revealed that SOX9 deletion affected BCL2 and survivin expression patterns, potentially explaining the altered apoptotic threshold [67].
Recent research has identified SOX9 as a critical mediator of resistance to combined anti-PD-1 and anti-LAG-3 immunotherapy in head and neck squamous cell carcinoma (HNSCC). Single-cell RNA sequencing of resistant tumors revealed significant enrichment of SOX9+ tumor cells that actively remodel the tumor microenvironment to confer treatment resistance [27].
Experimental Evidence: In a 4NQO-induced HNSCC mouse model, researchers demonstrated that SOX9+ epithelial tumor cells mediate apoptosis of Fpr1+ neutrophils through the Anxa1-Fpr1 axis [27]. This pathway promotes mitochondrial fission and inhibits mitophagy by suppressing Bnip3 expression, ultimately preventing neutrophil accumulation in tumor tissues [27]. The reduction of Fpr1+ neutrophils impairs infiltration and cytotoxic function of CD8+ T and γδT cells, creating an immunosuppressive microenvironment resistant to combination therapy [27]. This mechanism was validated through multiple transgenic mouse models, confirming SOX9's direct role in regulating Anxa1 transcription and immune evasion.
Table 2: SOX9 in Immunotherapy Resistance: Key Experimental Findings
| Cancer Type | Immunotherapy | Experimental Model | Resistance Mechanism | Validation Approach |
|---|---|---|---|---|
| Head and Neck Squamous Cell Carcinoma | Anti-LAG-3 + Anti-PD-1 | 4NQO-induced HNSCC mouse model | SOX9+ cells mediate Fpr1+ neutrophil apoptosis via Anxa1-Fpr1 axis | Transgenic mouse models; scRNA-seq |
| Various Cancers (Latent Metastasis) | Immune Surveillance | Metastatic dormancy models | SOX9 maintains stemness and immune evasion in dormant cells | In vivo imaging; immune cell depletion studies |
Beyond direct immunotherapy resistance, SOX9 significantly influences broader immune landscape organization. Bioinformatics analyses across multiple cancer types demonstrate strong correlations between SOX9 expression patterns and immune cell infiltration [4]. In colorectal cancer, SOX9 expression negatively correlates with infiltration of B cells, resting mast cells, resting T cells, monocytes, plasma cells, and eosinophils, while showing positive correlation with neutrophils, macrophages, activated mast cells, and naive/activated T cells [4]. Similarly, in prostate cancer, SOX9 expression is associated with an "immune desert" microenvironment characterized by decreased effector immune cells and increased immunosuppressive populations [4].
CRISPR/Cas9-Mediated Knockout: Multiple studies utilized CRISPR/Cas9 technology for complete SOX9 ablation. In the chondrosarcoma study, two single-cell-derived colonies (clones 8 and 11) with complete SOX9 knockout were selected and sequenced to verify generated mutations [67]. Both SOX9 alleles were disrupted by deletion or insertion, resulting in total SOX9 knockout confirmed via Western blotting [67]. These knockout clones showed morphological variations and were characterized for multiple cancer-related phenotypes.
siRNA-Mediated Knockdown: Transient SOX9 reduction via siRNA achieved 80-90% decrease in protein expression compared to control cells (non-targeting siRNA) [67]. The differential outcomes between complete knockout and partial knockdown suggest threshold effects for particular SOX9-mediated functions, highlighting the importance of selection between these technical approaches based on research objectives.
Single-Cell RNA Sequencing: Comprehensive scRNA-seq analysis of 51,786 cells from patient HGSOC tumors before and after platinum/taxane neoadjuvant chemotherapy identified 8,806 epithelial cancer cells and demonstrated consistent SOX9 upregulation post-treatment [17]. Analytical approaches included CopyKAT for distinguishing malignant from non-malignant cells and detailed subclustering to identify therapy-resistant populations [27].
Transcriptional Divergence Analysis: This metric, defined as the sum of expression of the top 50% of detected genes divided by the sum of expression of the bottom 50% (P50/P50), was employed to quantify transcriptional plasticity [17]. SOX9 expression strongly correlated with increased transcriptional divergence, indicating enhanced cellular plasticity and stemness characteristics [17].
Diagram 1: SOX9-Mediated Therapy Resistance Mechanisms. SOX9 drives resistance through dual pathways: enhancing cancer cell plasticity for chemotherapy resistance and remodeling the tumor microenvironment for immunotherapy resistance.
Table 3: Essential Research Reagents for SOX9 Functional Studies
| Reagent/Resource | Specific Examples | Application | Key Considerations |
|---|---|---|---|
| CRISPR/Cas9 Systems | SOX9-targeting sgRNA; Cas9 expression vectors | Complete SOX9 knockout | Verify biallelic disruption; single-cell clone selection |
| siRNA Oligonucleotides | Human SOX9-specific siRNA | Transient SOX9 knockdown | Achieves 80-90% protein reduction; suitable for acute effects |
| Cell Line Models | HGSOC: OVCAR4, Kuramochi, COV362; Chondrosarcoma: HTB94 | In vitro therapeutic testing | Context-dependent SOX9 functions; lineage-specific effects |
| Animal Models | 4NQO-induced HNSCC; Patient-derived xenografts | In vivo validation | Better recapitulates tumor microenvironment |
| scRNA-seq Platforms | 10X Genomics; CopyKAT analysis | Tumor heterogeneity analysis | Identifies rare SOX9+ subpopulations; cellular plasticity |
| SOX9 Antibodies | Validation for IHC, Western blotting | Expression analysis | Confirm specificity for intended applications |
The collective evidence from preclinical models solidifies SOX9's position as a master regulator of therapy resistance across multiple cancer types and treatment modalities. SOX9 knockout consistently sensitizes tumors to conventional chemotherapies like platinum agents and doxorubicin, while also reversing resistance to combination immunotherapies targeting PD-1 and LAG-3. The molecular mechanisms underlying these effects involve SOX9's dual functionality in regulating intrinsic cancer cell plasticity and stemness while simultaneously shaping an immunosuppressive tumor microenvironment.
Future research directions should focus on developing clinically viable SOX9 inhibition strategies, including small molecule inhibitors, degrader technologies, and combinatorial approaches with existing standard-of-care treatments. The context-dependent nature of SOX9 function, particularly its role as both oncogene and tumor suppressor in different malignancies, necessitates careful therapeutic development with appropriate patient stratification strategies. Nevertheless, targeting SOX9 represents a promising approach for overcoming therapeutic resistance and improving outcomes across multiple solid tumors.
The transcription factor SRY-box transcription factor 9 (SOX9) has emerged as a critical regulator of tumor progression and the tumor immune microenvironment. Recent evidence positions SOX9 as a promising predictive biomarker for immunotherapy response across multiple cancer types. This guide synthesizes current research comparing SOX9-driven resistance mechanisms with conventional immunotherapy outcomes, providing researchers and drug development professionals with experimental data and methodologies for evaluating SOX9 in therapeutic contexts.
SOX9 is a transcription factor containing a highly conserved high-mobility group (HMG) domain that recognizes specific DNA sequences and regulates gene transcription [15]. While crucial for normal developmental processes including chondrogenesis, sex determination, and organogenesis, SOX9 becomes dysregulated in numerous malignancies [3] [4].
Key oncogenic roles of SOX9 include:
Table 1: SOX9 Expression Patterns Across Cancer Types
| Cancer Type | SOX9 Expression | Correlation with Prognosis | Immune Context |
|---|---|---|---|
| Glioblastoma (GBM) | Highly expressed [15] [22] | Better prognosis in lymphoid invasion subgroups [15] | Correlated with immune infiltration [15] |
| Lung Cancer | Overexpressed [57] [20] | Poor survival [57] | Creates "immune cold" tumors [57] |
| Colorectal Cancer | Upregulated [4] | Poor prognosis [3] | Negative correlation with B cells, resting T cells [4] |
| Breast Cancer | Overexpressed [11] | Associated with basal-like subtype [11] | Promotes immune evasion [11] |
| Liver Cancer | Highly expressed [20] | Poor outcome [3] | Associated with immunosuppression [4] |
| Bone Tumors | Increased in malignant vs. benign [68] | Correlated with metastasis, recurrence [68] | Not specified |
| Melanoma | Decreased [20] | Tumor suppressor role [20] | Context-dependent |
SOX9 plays a multifaceted role in shaping the tumor immune microenvironment through several distinct mechanisms:
Immune Cell Infiltration: In multiple cancer types, SOX9 expression correlates with specific immune infiltration patterns. In colorectal cancer, SOX9 negatively correlates with infiltration of B cells, resting mast cells, resting T cells, monocytes, plasma cells, and eosinophils, while showing positive correlation with neutrophils, macrophages, activated mast cells, and naive/activated T cells [4]. Similarly, in lung cancer, SOX9 overexpression creates an "immune cold" environment characterized by poor immune cell infiltration [57].
Immune Checkpoint Regulation: SOX9 expression correlates with immune checkpoint expression in glioblastoma, suggesting its involvement in immunosuppressive pathways [15] [22]. Research in lung adenocarcinoma indicates SOX9 is mutually exclusive with various tumor immune checkpoints [15], pointing to complex regulatory relationships with existing immunotherapy targets.
Cellular Dormancy and Immune Evasion: Studies reveal that SOX9 enables dormant cancer cells to survive in secondary metastatic sites by avoiding immune surveillance, essentially creating a reservoir for future recurrence [11]. This immune evasion function positions SOX9 as a critical factor in metastatic progression and therapeutic resistance.
The following diagram illustrates the dual role of SOX9 in tumor immunity and its impact on immunotherapy response:
Evidence across multiple cancer types indicates that SOX9 expression levels may serve as a valuable biomarker for predicting response to conventional immunotherapies:
Lung Cancer: In KRAS-mutant lung cancer, SOX9 overexpression creates "immune cold" conditions where patients show poor response to immunotherapy [57]. This suggests testing Sox9 levels could identify patients unlikely to respond to immune checkpoint inhibitors.
Glioblastoma: Surprisingly, high SOX9 expression associates with better prognosis in specific lymphoid invasion subgroups, indicating context-dependent effects [15]. This highlights the need for cancer-type-specific evaluation of SOX9 as a biomarker.
Pan-Cancer Analysis: Comprehensive analysis of 33 cancer types reveals SOX9 overexpression in 15 cancers, with high expression correlating with worst overall survival in LGG, CESC, and THYM, supporting its utility as a prognostic marker [20].
Table 2: SOX9 Inhibition vs. Conventional Immunotherapy Mechanisms
| Therapeutic Approach | Mechanism of Action | Resistance Mechanisms | Biomarker Potential |
|---|---|---|---|
| SOX9 Inhibition | Direct targeting of tumor stemness and immune evasion at transcriptional level [3] [11] | Alternative signaling pathway activation; SOX9 protein stability [3] | SOX9 expression levels predict response; downstream target expression [57] |
| Immune Checkpoint Inhibitors | Reactivating T-cell mediated tumor cell killing [4] | SOX9-mediated T-cell exclusion; immunosuppressive microenvironment [57] | PD-L1 expression; tumor mutational burden; SOX9 as negative predictor [57] |
| Combination Therapy | SOX9 inhibition to reverse "immune cold" phenotype followed by checkpoint blockade [57] | Complex feedback mechanisms; toxicity concerns | Dynamic monitoring of immune cell infiltration; SOX9 downstream targets [15] |
Lung Cancer Models: Research demonstrates that Sox9 knockout delays tumor formation, while Sox9 overexpression accelerates tumor development in KRAS-driven lung cancer [57]. This effect was primarily mediated through profound impacts on immune cell infiltration.
Liver Fibrosis Models: In mouse models of liver fibrosis, ablation of SOX9 significantly reduced scarring, improved liver function, and decreased inflammation [69]. This demonstrates SOX9's fundamental role in fibrotic processes that can shape the tumor microenvironment.
Bone Cancer Clinical Correlation: In human bone tumor samples, SOX9 overexpression correlates with tumor severity, grade, invasion features, poor response to therapy, and recurrence [68]. This clinical evidence supports SOX9's role in therapeutic resistance.
Transcriptomic Analysis of SOX9-Related Immune Signatures:
Immune Infiltration Analysis:
Functional Validation Experiments:
The following workflow diagram outlines a comprehensive approach to evaluating SOX9 as an immunotherapy biomarker:
Table 3: Key Research Reagents for SOX9 Studies
| Reagent/Category | Specific Examples | Research Application | Considerations |
|---|---|---|---|
| SOX9 Detection | Anti-SOX9 antibodies (IHC, WB); SOX9 ELISA kits [68] [69] | Protein expression quantification in tissues and serum | Validate antibody specificity; establish appropriate controls |
| Gene Expression Analysis | SOX9 siRNA/shRNA; CRISPR-Cas9 systems; RT-PCR primers [69] [20] | Functional studies of SOX9 manipulation | Optimize knockdown efficiency; control for off-target effects |
| Cell Line Models | Prostate: 22RV1, PC3; Lung: H1975; Various cancer cell lines [20] | In vitro mechanistic studies | Select appropriate models reflecting cancer type specificity |
| Small Molecule Inhibitors | Cordycepin (adenosine analog) [20] | SOX9 pathway inhibition studies | Dose-response optimization; specificity validation |
| Database Resources | HPA, TCGA, GTEx, cBioPortal, GEPIA2 [15] [20] | Bioinformatics analysis of SOX9 in human cancers | Use consistent normalization methods across datasets |
SOX9 represents a promising predictive biomarker for immunotherapy response with context-dependent effects across different cancer types. The current evidence supports:
The dual role of SOX9 in both tumor progression and immune regulation positions it uniquely at the intersection of tumor-intrinsic and tumor-extrinsic pathways, offering exciting opportunities for therapeutic intervention beyond conventional immunotherapy approaches.
The transcription factor SOX9 (SRY-box 9) has emerged as a critical driver of therapy resistance across multiple cancer types, a finding increasingly elucidated through single-cell RNA sequencing (scRNA-seq) technologies. Resistance to chemotherapy, targeted therapy, and immunotherapy remains a formidable challenge in oncology, and understanding the molecular mechanisms underlying this process is essential for developing effective countermeasures. Single-cell transcriptomics has revealed that SOX9-expressing tumor cells frequently survive treatment and initiate relapse, positioning SOX9 as a central player in treatment failure. This review synthesizes evidence from recent scRNA-seq studies demonstrating SOX9 enrichment in resistant tumors and compares the effectiveness of targeting SOX9-driven resistance pathways versus conventional immunotherapeutic approaches.
In high-grade serous ovarian cancer (HGSOC), scRNA-seq of patient tumors before and after platinum/taxane neoadjuvant chemotherapy revealed that SOX9 is significantly upregulated in post-treatment cancer cells [17]. Analysis of 8,806 epithelial cancer cells from 11 patients showed SOX9 expression consistently increased after chemotherapy in 8 of the 11 patients, both at single-cell and patient-specific pseudo-bulk RNA levels [17]. This chemotherapy-induced SOX9 upregulation was sufficient to reprogram naive ovarian cancer cells into a stem-like state characterized by increased transcriptional divergenceâa metric of transcriptional plasticity amplified in stem and cancer stem cells (CSCs) [17].
Table 1: SOX9 in Chemotherapy-Resistant Ovarian Cancer
| Evidence Type | Finding | Experimental Support |
|---|---|---|
| scRNA-seq Analysis | SOX9 significantly upregulated in post-chemotherapy tumor cells | 51,786 cells analyzed from 11 HGSOC patients pre/post NACT [17] |
| Functional Association | SOX9 induces stem-like transcriptional state and platinum resistance | CRISPR/Cas9 knockout increased carboplatin sensitivity (P=0.0025) [17] |
| Clinical Correlation | High SOX9 expression associated with poorer overall survival | HR=1.33; log-rank P=0.017 for top vs bottom SOX9 quartiles [17] |
| Mechanistic Insight | SOX9 increases transcriptional divergence (P50/P50 metric) | Indicator of enhanced transcriptional plasticity and stemness [17] |
In head and neck squamous cell carcinoma (HNSCC), scRNA-seq of mouse models resistant to anti-LAG-3 plus anti-PD-1 combination immunotherapy revealed significant enrichment of SOX9+ tumor cells [27]. Analysis of 19,917 malignant cells identified distinct epithelial subclusters, with SOX9-high populations preferentially expanded in treatment-resistant tumors [27]. Mechanistically, SOX9 was found to directly regulate annexin A1 (Anxa1) expression, which mediated apoptosis of formyl peptide receptor 1 (Fpr1)+ neutrophils through the Anxa1-Fpr1 axis, ultimately preventing neutrophil accumulation and impairing cytotoxic CD8+ T and γδT cell infiltration and function [27].
Table 2: SOX9 in Immunotherapy-Resistant Head and Neck Cancer
| Evidence Type | Finding | Experimental Support |
|---|---|---|
| scRNA-seq Analysis | SOX9+ tumor cells enriched in anti-LAG-3/PD-1 resistant tumors | 19,917 malignant cells from HNSCC mouse model [27] |
| Resistance Rate | 42.9% of animals resistant to combination therapy | 6 of 14 mice showed tumor growth despite treatment [27] |
| Mechanistic Pathway | SOX9âANXA1âFPR1 neutrophil axis reduces cytotoxic T cells | SOX9 transcriptionally regulates ANXA1, causing FPR1+ neutrophil apoptosis [27] |
| Immune Context | Resistant tumors showed reduced immune cell infiltration | Proportion of immune cells dramatically lower in resistant vs sensitive group [27] |
In esophageal cancer (ESCA), scRNA-seq analysis identified SOX9 as a key marker of cancer stem cells (CSCs), with SOX9-expressing cells demonstrating enhanced stemness potential as measured by CytoTRACE algorithm [70]. These SOX9+ CSCs contributed significantly to tumor heterogeneity and therapeutic resistance, forming a basis for developing prognostic models of treatment response [70].
The standard scRNA-seq workflow for identifying SOX9-enriched resistant populations involves:
Sample Processing: Fresh tumor tissues are dissociated into single-cell suspensions using enzymatic digestion (collagenase/hyaluronidase mixtures) followed by mechanical dissociation [27].
Cell Viability and Quality Control: Viable cell counts are determined using trypan blue exclusion or automated cell counters, with targets of >80% viability before sequencing [71].
Single-Cell Partitioning: Cells are partitioned into nanoliter-scale droplets using microfluidic devices (10X Genomics Chromium system) where each cell is barcoded with a unique molecular identifier (UMI) [71].
Library Preparation and Sequencing: Reverse transcription, cDNA amplification, and library construction are performed following manufacturer protocols, with sequencing typically on Illumina platforms to depth of 50,000-100,000 reads per cell [71] [27].
Bioinformatic Analysis:
SOX9 Knockout Models: CRISPR/Cas9-mediated SOX9 knockout using lentiviral delivery of SOX9-targeting sgRNA, with validation of knockout efficiency via Western blot and qRT-PCR [17].
In Vivo Treatment Resistance Models:
Stemness Assays: Colony formation assays to assess self-renewal capability; flow cytometry analysis of stem cell markers (CD44, CD133, ALDH) in SOX9-high versus SOX9-low populations [17].
Diagram 1: SOX9-Driven Resistance Pathways. SOX9 activation by multiple therapy types promotes resistance through stem cell reprogramming leading to both cell-intrinsic (transcriptional divergence) and microenvironmental (ANXA1-FPR1 neutrophil axis) mechanisms.
Despite the efficacy of immune checkpoint inhibitors (anti-PD-1, anti-PD-L1, anti-LAG-3) in treating various cancers, a substantial proportion of patients develop resistance through multiple mechanisms [27] [72]. In HNSCC, even combination therapy with anti-LAG-3 plus anti-PD-1 fails in approximately 43% of cases, with SOX9+ tumor cells identified as key mediators of this resistance [27]. Similarly, in breast cancer, resistance to CDK4/6 inhibitors frequently develops through heterogeneous mechanisms that may involve SOX9-related pathways [73] [74].
Targeting SOX9 and its downstream effectors represents a promising alternative or complementary strategy to conventional immunotherapy:
Direct SOX9 Targeting: CRISPR/Cas9-mediated SOX9 knockout significantly sensitized ovarian cancer cells to platinum-based chemotherapy (P=0.0025), demonstrating the potential of direct SOX9 inhibition [17].
ANXA1-FPR1 Axis Disruption: In HNSCC, blocking the SOX9-ANXA1-FPR1 pathway prevented neutrophil apoptosis and restored cytotoxic T cell function, potentially overcoming resistance to combination immunotherapy [27].
Stemness Pathway Inhibition: Targeting SOX9-driven stem-like properties may prevent the emergence of treatment-resistant CSC populations that drive tumor recurrence [17] [70].
Table 3: SOX9 Inhibition vs Conventional Immunotherapy
| Therapeutic Approach | Mechanism of Action | Resistance Challenges | Potential Synergy with SOX9 Inhibition |
|---|---|---|---|
| Anti-PD-1/PD-L1 | Blocks T-cell inhibitory signals | SOX9-mediated T-cell exclusion via neutrophil apoptosis [27] | SOX9 inhibition restores neutrophil-mediated T-cell recruitment |
| Anti-LAG-3 | Reverses T-cell exhaustion | SOX9+ tumor cells evade despite combination therapy [27] | Prevents SOX9-driven adaptive resistance |
| CDK4/6 Inhibitors | Cell cycle arrest in ER+ breast cancer | Heterogeneous resistance mechanisms [73] [74] | SOX9 inhibition targets resistant stem-like populations |
| Chemotherapy | DNA damage and cell death | SOX9-induced stem-like state with enhanced DNA repair [17] | SOX9 knockout increases chemosensitivity |
Table 4: Key Research Reagents for SOX9 Resistance Studies
| Reagent/Category | Specific Examples | Research Application | Key Function |
|---|---|---|---|
| scRNA-seq Platforms | 10X Genomics Chromium, Smart-seq2 | Transcriptomic profiling | Single-cell resolution of SOX9+ subpopulations [71] |
| SOX9 Detection | Anti-SOX9 antibodies, SOX9 FISH probes | Protein/DNA localization | Identify SOX9+ cells in tissue context [17] |
| Animal Models | 4NQO-induced HNSCC, Patient-derived xenografts | In vivo resistance studies | Model therapy response and resistance [27] |
| Gene Editing | CRISPR/Cas9 SOX9 sgRNA, Lentiviral vectors | Functional validation | SOX9 knockout to assess necessity [17] |
| Cell Sorting | FACS antibodies (CD45, EPCAM, SOX9 reporters) | Population isolation | Purify SOX9+ vs SOX9- cells for functional assays |
| Pathway Inhibitors | ANXA1 inhibitors, FPR1 antagonists | Mechanistic studies | Block specific SOX9 effector pathways [27] |
Single-cell RNA sequencing evidence consistently demonstrates SOX9 enrichment across multiple therapy-resistant cancer types, establishing SOX9 as a central node in treatment failure. SOX9 drives resistance through dual mechanisms: intrinsically, by promoting a stem-like state with enhanced transcriptional plasticity; and extrinsically, by remodeling the tumor immune microenvironment to exclude cytotoxic immune cells. Targeting SOX9 and its downstream effectors represents a promising strategy to overcome limitations of conventional immunotherapy, particularly in treatment-resistant settings. Future research should focus on developing clinical-grade SOX9 inhibitors, validating SOX9-based patient stratification biomarkers, and designing rational combination therapies that simultaneously target SOX9 pathways and conventional immune checkpoints.
The evolving landscape of oncology increasingly focuses on targeted therapies and immunomodulatory approaches to combat treatment resistance and improve patient outcomes. Two distinct strategic paradigms have emerged: transcription factor targeting, exemplified by SOX9 inhibition, and immune checkpoint blockade (ICB) monotherapy, which modulates the tumor microenvironment (TME). SOX9, a transcription factor critical in embryonic development and stem cell maintenance, has been identified as a significant oncogenic driver in multiple cancer types, where it promotes tumor initiation, stemness, and metastasis [22] [31]. Conversely, ICB monotherapy, particularly targeting PD-1/PD-L1 pathways, aims to reactivate suppressed anti-tumor immune responses, though its efficacy varies substantially across cancer types and molecular subtypes [75] [76]. This comparison guide provides a systematic analysis of the therapeutic outcomes, molecular mechanisms, and clinical applications of these divergent approaches, contextualizing their respective roles in precision oncology.
SOX9 exerts its oncogenic functions through multifaceted mechanisms that sustain tumorigenesis and therapeutic resistance. In glioblastoma (GBM), SOX9 demonstrates significantly elevated expression in tumor tissues compared to normal brain, functioning as a diagnostic and prognostic biomarker, particularly in isocitrate dehydrogenase (IDH)-mutant cases [22]. High-throughput transcriptional analyses have revealed that SOX9 regulates a cancer-specific gene network that promotes stemness, extracellular matrix (ECM) deposition, and cytoskeleton remodeling while simultaneously repressing cellular differentiation programs [31]. This maintenance of cancer stem cell populations enables long-term tumor propagation and resistance to conventional therapies.
In the context of immune regulation, SOX9 expression demonstrates significant correlation with immune cell infiltration patterns and immune checkpoint expression in the TME [22]. Recent investigations in head and neck squamous cell carcinoma (HNSCC) models resistant to combined anti-PD-1 and anti-LAG-3 therapy have identified a novel resistance mechanism mediated by SOX9+ tumor cells. These cells directly regulate annexin A1 (Anxa1) expression, which subsequently initiates apoptosis of formyl peptide receptor 1 (Fpr1)+ neutrophils through the Anxa1-Fpr1 axis. This pathway promotes mitochondrial fission and inhibits mitophagy by downregulating BCL2/adenovirus E1B interacting protein 3 (Bnip3) expression, ultimately preventing neutrophil accumulation in tumor tissues [27]. The consequent reduction of Fpr1+ neutrophils impairs the infiltration and cytotoxic capacity of CD8+ T and γδT cells within the TME, establishing an immunosuppressive niche that facilitates therapy resistance.
Immune checkpoint blockade monotherapy primarily targets inhibitory receptors on immune cells, notably PD-1, CTLA-4, and LAG-3, to restore anti-tumor immunity. These checkpoints normally function as physiological brakes on immune activation to prevent autoimmunity, but tumors co-opt these pathways to evade immune surveillance [76]. PD-1 inhibitors disrupt the interaction between PD-1 on T cells and PD-L1/PD-L2 on tumor and immune cells, reversing T-cell exhaustion and restoring cytotoxic function [75] [27].
The efficacy of ICB monotherapy is profoundly influenced by specific oncogenic mutations and TME composition. In metastatic lung adenocarcinoma (LUAD), patients with KRAS mutations (particularly G12C and G12V subtypes) derive substantial benefit from ICB monotherapy, demonstrating significantly improved overall survival (16 months versus 8 months) and progression-free survival (8 months versus 5 months) compared to chemotherapy [75] [77]. Conversely, KRAS wild-type patients show no survival advantage from ICB monotherapy, though they may benefit from chemoimmunotherapy combinations [75]. This mutation-specific response highlights the importance of molecular stratification in ICB treatment decisions.
The TME significantly modulates ICB responses through multiple resistance mechanisms, including immune checkpoint upregulation, antigen loss or downregulation, T-cell exhaustion, and the presence of immunosuppressive cells (Tregs, MDSCs) and cytokines (TGF-β, IL-10) [76]. Hypoxic conditions within the TME further promote resistance by enhancing PD-L1 expression and facilitating glycolytic metabolism through lactylation of SOX9, creating a feedback loop that reinforces immunosuppression [76].
Figure 1: Comparative Molecular Mechanisms of SOX9 Inhibition and Immune Checkpoint Blockade. SOX9 promotes tumor progression through stemness maintenance, ECM remodeling, and neutrophil-mediated immunosuppression via the ANXA1-FPR1 axis. Immune checkpoint blockade reactivates suppressed T-cell function through PD-1, CTLA-4, and LAG-3 inhibition.
Table 1: Therapeutic Outcomes Across Cancer Types
| Cancer Type | Therapeutic Approach | Primary Outcomes | Molecular Context | Study Details |
|---|---|---|---|---|
| Glioblastoma | SOX9 High Expression | Diagnostic & prognostic biomarker; Better prognosis in lymphoid invasion subgroups (p<0.05) | IDH-mutant cases; Associated with immune infiltration | 478 cases; RNA-seq from TCGA/GTEx [22] |
| Lung Adenocarcinoma | ICB Monotherapy | OS: 16 vs 8 months (p<0.001); PFS: 8 vs 5 months (p<0.001) | KRAS-mutant (G12C/G12V) | 424 patients; Retrospective cohort [75] [77] |
| Lung Adenocarcinoma | ICB Monotherapy | No survival advantage (OS: 8 vs 8 months, p=0.648) | KRAS wild-type | 424 patients; Retrospective cohort [75] |
| Head & Neck SCC | SOX9+ Tumor Cells | Mediates resistance to anti-LAG-3+anti-PD-1 therapy | Regulates ANXA1-FPR1 neutrophil axis | scRNA-seq of resistant tumors [27] |
| Colorectal Cancer | SOX9 Inhibition | Suppresses tumor growth via EMT inhibition | Suppresses SOX2 induction | Preclinical models [9] |
| Multiple Solid Tumors | Dual ICB (anti-CTLA4+anti-PD1) | Prolonged PFS: 34-69 months | Includes MSS colorectal, neuroendocrine, prostate | Case series (n=5) [78] |
Table 2: Resistance Mechanisms and Biomarkers
| Resistance Mechanism | SOX9 Inhibition Context | ICB Monotherapy Context | Potential Overcoming Strategies |
|---|---|---|---|
| Immune Cell Modulation | Reduces Fpr1+ neutrophil accumulation via ANXA1-FPR1 axis [27] | T-cell exhaustion; Immunosuppressive TME (Tregs, MDSCs) [76] | Neutrophil-targeted combinations; TME reprogramming |
| Metabolic Adaptation | Not well characterized | Hypoxia-induced glycolysis via SOX9 lactylation [76] | Metabolic interventions; HIF inhibitors |
| Stemness Maintenance | Promotes cancer stem cell self-renewal [31] | Cancer stem cell persistence despite ICB | Dual targeting of stemness and immune pathways |
| Checkpoint Adaptation | Correlated with immune checkpoint expression [22] | Upregulation of alternative checkpoints (TIM-3, LAG-3) [76] | Multi-checkpoint inhibition; Sequential targeting |
| Mutation-Specific Responses | Independent prognostic factor in IDH-mutant GBM [22] | KRAS mutation status predicts ICB benefit in LUAD [75] | Molecular profiling for patient selection |
Comprehensive characterization of SOX9 function in cancer employs multi-omics approaches and sophisticated genetic models. Standard protocols include:
RNA Sequencing and Differential Expression Analysis: RNA-seq data for SOX9 expression analysis is typically obtained from large-scale databases such as The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx). Data processing utilizes HTSeq-FPKM and HTSeq-Count formats, with differential expression analysis performed using DESeq2 R package with thresholds of |log fold change (logFC)| >2 and adjusted p-value <0.05 [22]. Functional enrichment analysis of SOX9-correlated genes is conducted through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and Gene Set Enrichment Analysis (GSEA) using ClusterProfiler R package with 1,000 permutations [22] [15].
Protein-Protein Interaction Network Mapping: Differentially expressed genes are analyzed using the Search Tool for the Retrieval of Interacting Genes (STRING) database with an interaction score threshold of 0.4. Resulting networks are visualized and analyzed using Cytoscape with Molecular Complex Detection (MCODE) plugin to identify significant functional modules (MCODE scores >5, degree cut-off=2, node score cut-off=0.2) [22].
Genetic Mouse Models: Conditional knockout models (e.g., Sox9fl/fl:AlbCre mice) enable tissue-specific deletion of Sox9 beginning at E10.5, permitting analysis of its role in tumor initiation and progression [79]. Phenotypic characterization employs large-volume 3D imaging techniques (iDISCO+ and light sheet microscopy) with quantitative morphological analysis using Sholl methodology to quantify branching complexity in structures such as intrahepatic bile ducts [79].
Single-Cell RNA Sequencing for Resistance Mechanisms: Analysis of therapy-resistant tumors involves single-cell dissociation followed by 10X Genomics platform library construction. Cell clustering identifies distinct subpopulations, while CopyKAT analysis distinguishes malignant from non-malignant cells. Cellular interactions are deciphered through ligand-receptor pairing tools such as CellPhoneDB [27].
Standardized methodologies for evaluating ICB efficacy include:
Retrospective Clinical Cohort Analysis: Multicenter studies collect comprehensive patient data including demographics, tumor characteristics, treatment details, and survival outcomes from cancer registries and medical charts. KRAS mutational status is determined by next-generation sequencing. Survival analysis employs Kaplan-Meier curves with log-rank tests and multivariate Cox regression models to calculate hazard ratios with 95% confidence intervals [75] [77].
Immune Cell Infiltration Quantification: Computational algorithms including ssGSEA (single-sample Gene Set Enrichment Analysis) and ESTIMATE are applied to RNA-seq data to infer immune cell abundances within the TME. Correlation analyses between SOX9 expression and immune infiltration utilize Spearman's test and Wilcoxon rank sum test [22].
Response Evaluation: Tumor response is assessed according to Response Evaluation Criteria in Solid Tumors (RECIST) guidelines, with resistant tumors defined as those showing >20% increase in size post-treatment [27]. Immunological monitoring includes flow cytometric analysis of immune cell populations and immunohistochemical staining for proliferation markers (Ki-67) and apoptosis markers (cleaved caspase-3) [27].
Figure 2: Integrated Experimental Workflow for SOX9 and Immune Checkpoint Blockade Research. Comprehensive analysis combines molecular profiling, computational biology, functional validation in models, and clinical correlation studies.
Table 3: Essential Research Reagents and Resources
| Reagent/Resource | Primary Application | Specific Function | Example Implementation |
|---|---|---|---|
| TCGA/GTEx Databases | Transcriptomic analysis | SOX9 expression profiling across cancers | Pan-cancer RNA-seq data analysis [22] |
| DESeq2 R Package | Bioinformatics | Differential expression analysis | Identifying SOX9-correlated genes [22] |
| STRING Database | Network biology | Protein-protein interaction mapping | Constructing SOX9-regulated networks [22] |
| Cytoscape with MCODE | Network visualization | Module identification in PPI networks | Analyzing SOX9 functional modules [22] |
| ssGSEA/ESTIMATE | Immunogenomics | Immune cell infiltration quantification | Correlating SOX9 with immune profiles [22] |
| Sox9-floxed Mice | Genetic modeling | Conditional Sox9 knockout studies | BCC formation and IHBD development [79] [31] |
| Anti-PD-1/Anti-LAG-3 | Immunotherapy research | Immune checkpoint blockade studies | Resistance mechanism analysis [27] |
| Single-Cell RNA Seq | Cellular heterogeneity | Tumor subpopulation identification | Resistant cell characterization [27] |
| Sholl Analysis | Morphometric analysis | Branching complexity quantification | IHBD morphological assessment [79] |
| ClusterProfiler | Functional genomics | GO/KEGG pathway enrichment | SOX9-related pathway analysis [22] |
The comparative analysis of SOX9 inhibition and immune checkpoint blockade monotherapy reveals two fundamentally distinct yet potentially complementary approaches to cancer treatment. SOX9 inhibition targets core oncogenic processesâstemness maintenance, differentiation blockade, and immune evasionâmaking it particularly relevant in IDH-mutant glioblastoma and other malignancies where SOX9 drives tumor initiation and progression [22] [31]. Conversely, ICB monotherapy demonstrates remarkable efficacy in specific molecular contexts, most notably KRAS-mutant lung adenocarcinoma, but suffers from limited response rates in unselected populations and acquired resistance mechanisms [75] [76].
The emerging understanding of SOX9's role in modulating the tumor immune microenvironment, particularly through the newly identified ANXA1-FPR1 neutrophil axis, suggests potential synergistic opportunities for combination approaches [27]. Furthermore, the observation that hypoxia-induced SOX9 lactylation contributes to immunosuppressive TME formation indicates possible feedback loops between these pathways [76]. Future research directions should prioritize the development of specific SOX9 inhibitors, validation of combinatorial approaches targeting both oncogenic transcription factors and immune checkpoints, and refinement of patient selection strategies based on integrated molecular profiling encompassing both tumor-intrinsic pathways and immune microenvironment characteristics.
The SRY-box transcription factor 9 (SOX9) has emerged as a critical regulator of cancer progression and therapeutic resistance across diverse malignancies. While initially recognized for its roles in embryonic development and chondrogenesis, SOX9 is frequently dysregulated in human cancers, where it influences key oncogenic processes including cell proliferation, stemness, epithelial-mesenchymal transition (EMT), and drug resistance [20] [3]. More recently, compelling evidence has revealed that SOX9 also serves as a master regulator of tumor-immune interactions, shaping an immunosuppressive microenvironment that limits the efficacy of immunotherapies [4] [27]. This review systematically compares the therapeutic potential of targeting SOX9 in combination with established immunotherapeutic regimens, particularly immune checkpoint inhibitors (ICIs), against conventional immunotherapy approaches alone. We synthesize current preclinical evidence, elucidate underlying mechanisms, and provide detailed experimental methodologies to guide future research and drug development efforts in this emerging field.
Table 1: SOX9 Expression and Prognostic Significance Across Cancers
| Cancer Type | SOX9 Expression Pattern | Correlation with Immune Parameters | Prognostic Value |
|---|---|---|---|
| Glioblastoma (GBM) | Highly expressed | Correlated with immune cell infiltration and checkpoint expression | Better prognosis in lymphoid invasion subgroups [15] |
| Head and Neck Squamous Cell Carcinoma (HNSCC) | Enriched in therapy-resistant cells | Mediates neutrophil apoptosis via ANXA1-FPR1 axis | Associated with anti-PD-1/anti-LAG-3 resistance [27] |
| Breast Cancer | Frequently overexpressed | Triggers tumorigenesis via immune escape; maintains stemness | Driver of basal-like subtype [4] [11] |
| Lung Cancer | Significantly upregulated | Suppresses TME; mutually exclusive with immune checkpoints | Correlates with poorer overall survival [20] [15] |
| Colorectal Cancer | Highly expressed | Negatively correlates with B cells, resting mast cells, monocytes | Poor prognosis [4] [3] |
| Melanoma | Decreased expression | Inhibition of SOX9 restores immune sensitivity | Functions as tumor suppressor in this context [20] |
SOX9 modulates tumor-immune interactions through multiple interconnected mechanisms that collectively foster an immunosuppressive landscape and enable immunotherapy resistance.
Bioinformatics analyses of tumor transcriptomes have revealed striking correlations between SOX9 expression and specific immune infiltration patterns. In colorectal cancer, SOX9 expression negatively correlates with infiltration levels of B cells, resting mast cells, resting T cells, monocytes, plasma cells, and eosinophils, while showing positive correlations with neutrophils, macrophages, and activated mast cells [4]. Similarly, in breast cancer, SOX9 overexpression triggers tumorigenesis by facilitating the immune escape of tumor cells [20] [11]. A particularly crucial mechanism was identified in head and neck squamous cell carcinoma (HNSCC), where SOX9 directly regulates the expression of annexin A1 (Anxa1), mediating apoptosis of formyl peptide receptor 1 (Fpr1)+ neutrophils through the Anxa1-Fpr1 axis [27]. This pathway promotes mitochondrial fission, inhibits mitophagy by downregulating BCL2/adenovirus E1B interacting protein 3 (Bnip3) expression, and ultimately prevents neutrophil accumulation in tumor tissues. The subsequent reduction of Fpr1+ neutrophils impairs the infiltration and tumor cell-killing capacity of cytotoxic CD8+ T and γδT cells within the tumor microenvironment, thereby driving resistance to combination immunotherapy [27].
Beyond regulating immune cell recruitment, SOX9 engages in direct molecular crosstalk with established immune checkpoint systems. In breast cancer, SOX9 establishes a molecular axis with the immune checkpoint molecule B7x (B7-H4/VTCN1), creating a protective shield around dedifferentiated tumor cells that safeguards them from immune surveillance [80]. This SOX9-B7x axis represents a novel immune evasion mechanism that drives breast cancer progression independent of canonical PD-1/PD-L1 pathways. Additionally, analyses in glioblastoma and lung adenocarcinoma have revealed significant correlations between SOX9 expression and various tumor immune checkpoints, suggesting coordinated regulation of multiple immunosuppressive pathways [15].
Figure 1: SOX9-Mediated Immunosuppressive Mechanisms. SOX9 drives immunotherapy resistance through multiple pathways including neutrophil apoptosis via ANXA1-FPR1 axis, B7x upregulation, immune checkpoint modulation, and cancer stemness maintenance.
Emerging preclinical evidence demonstrates that SOX9 targeting can fundamentally reshape the tumor immune microenvironment and overcome key resistance mechanisms that limit conventional immunotherapy.
Recent investigation using a HNSCC mouse model demonstrated that Sox9+ tumor cells mediate resistance to anti-LAG-3 plus anti-PD-1 combination therapy [27]. Single-cell RNA sequencing of resistant tumors revealed significant enrichment of Sox9+ tumor cells, which orchestrate immunosuppression through the Anxa1-Fpr1 axis leading to impaired neutrophil and T cell function. This mechanism explains why even advanced combination checkpoint blockade strategies often fail against SOX9-high tumors. Importantly, genetic or pharmacological targeting of SOX9 in this context reversed neutrophil apoptosis, restored cytotoxic T cell infiltration, and sensitized tumors to combination immunotherapy, demonstrating a potential strategy to overcome this resistance pathway [27].
The integration of SOX9 inhibition with immunotherapy demonstrates superior efficacy compared to monotherapy approaches across multiple cancer types. In triple-negative breast cancer (TNBC), c-Myc influences CDK inhibitor efficacy through modulation of the SOX9âFOXC1 transcriptional axis [81]. As c-Myc inhibition enhances PD-L1 blockade efficacy in murine models, and given the established regulatory relationship between c-Myc and SOX9, these findings suggest that SOX9 targeting may similarly potentiate immune checkpoint inhibition. Furthermore, in various cancer cell lines, cordycepin (CD), an adenosine analog, inhibited both protein and mRNA expressions of SOX9 in a dose-dependent manner, indicating its anticancer roles likely via SOX9 inhibition [20]. This pharmacological approach to SOX9 suppression provides a promising combination strategy with immunotherapy.
Table 2: Experimental Models of SOX9 Inhibition with Immunotherapy
| Cancer Model | SOX9 Targeting Approach | Combination Immunotherapy | Key Outcomes | Reference |
|---|---|---|---|---|
| HNSCC mouse model | Genetic manipulation | anti-LAG-3 + anti-PD-1 | Restored Fpr1+ neutrophil accumulation and CD8+ T cell cytotoxicity [27] | |
| Prostate cancer cells (22RV1, PC3) | Cordycepin (10-40 μM) | Not tested | Dose-dependent inhibition of SOX9 mRNA and protein [20] | |
| Lung cancer cell (H1975) | Cordycepin (10-40 μM) | Not tested | Dose-dependent inhibition of SOX9 mRNA and protein [20] | |
| Breast cancer models | c-Myc knockdown (regulates SOX9) | PD-L1 blockade | Enhanced tumor suppression and TME remodeling [81] | |
| Melanoma models | SOX9 upregulation | Not specified | Inhibited tumorigenicity; restored retinoic acid sensitivity [20] |
Robust experimental methodologies are essential for investigating the synergistic potential of SOX9 inhibition with immunotherapies and validating combinatorial efficacy.
Cell Culture and Treatment Protocols: Prostate cancer cells (PC3, 22RV1) and lung cancer cells (H1975) are maintained in RPMI 1640 or DMEM medium supplemented with 10-15% fetal bovine serum and 1% penicillin/streptomycin at 37°C with 5% CO2 [20]. For SOX9 inhibition studies, cells are inoculated in 12-well plates and treated with compounds such as cordycepin at final concentrations of 0, 10, 20, and 40 μM for 24 hours. Following treatment, protein and RNA are collected for Western blot and quantitative PCR analysis to assess SOX9 expression at both protein and transcriptional levels [20].
Western Blot Analysis: Cells are lysed in EBC buffer and 2ÃSDS loading buffer to collect proteins. Protein samples are boiled at 100°C for 5 minutes, electrophoresed in SDS-PAGE gels, and transferred to PVDF membranes under ice bath conditions. After transfer, membranes are blocked and incubated with appropriate primary and secondary antibodies for SOX9 detection, with visualization performed using enhanced chemiluminescence [20].
Animal Model Establishment: For HNSCC studies, C57BL/6 wild-type mice are fed with 4-nitroquinoline 1-oxide (4NQO) water for 16 weeks, followed by normal water for another 8 weeks to induce tumor formation [27]. Mice with similar-sized tumor lesions are then randomly divided into control and treatment groups.
Therapy Response Assessment: Tumor-bearing mice are treated with control IgG, anti-PD-1, anti-LAG-3, or anti-LAG-3 plus anti-PD-1 antibodies, with assessment every 4 days from initial treatment [27]. Based on RECIST criteria, tumors growing more than 20% larger than original size after 14 days are classified as resistant. Magnetic resonance imaging (MRI) provides non-invasive monitoring of tumor growth processes throughout the study.
Single-Cell RNA Sequencing Analysis: For comprehensive characterization of therapy resistance mechanisms, tumor tissues from control, resistant, and sensitive groups are dissected, pooled, and digested into single-cell suspensions [27]. After quality control and filtering, cells are subjected to scRNA-seq using standard platforms. Cell types are identified based on canonical markers: epithelial cells (Krt14, Krt5, Krt6a), fibroblasts (Col1a1, Col3a1, Apod), endothelial cells (Flt1, Pecam1, Eng), immune cells (Ptprc, Cd74, Cd3g), and muscle cells (Myl9, Myh11, Mylk). Malignant epithelial subpopulations are distinguished using CopyKAT analysis [27].
Figure 2: Experimental Workflow for Evaluating SOX9 Inhibition with Immunotherapy. Comprehensive methodology spanning in vitro SOX9 modulation to in vivo validation using murine cancer models and single-cell analysis.
Table 3: Key Research Reagents for Investigating SOX9 in Cancer Immunotherapy
| Reagent/Category | Specific Examples | Research Application | Function |
|---|---|---|---|
| SOX9 Inhibitors | Cordycepin | In vitro and in vivo SOX9 suppression | Adenosine analog that inhibits SOX9 mRNA and protein expression [20] |
| Immune Checkpoint Inhibitors | Anti-PD-1, Anti-LAG-3 | Combination therapy studies | Block inhibitory receptors on T cells to restore anti-tumor immunity [27] |
| Cell Lines | 22RV1, PC3, H1975 | In vitro mechanistic studies | Prostate and lung cancer models for SOX9 expression and inhibition studies [20] |
| Animal Models | 4NQO-induced HNSCC (C57BL/6) | In vivo therapy evaluation | Immunocompetent mouse model for studying immunotherapy resistance [27] |
| Analysis Tools | Single-cell RNA sequencing | Tumor microenvironment characterization | Identification of SOX9+ subpopulations and immune cell dynamics [27] |
| Antibodies | Anti-SOX9, Anti-Ki67, Anti-cleaved Caspase-3 | Immunohistochemistry and Western blot | Detection of SOX9 expression, proliferation, and apoptosis [20] [27] |
The accumulating evidence firmly establishes SOX9 as a critical regulator of tumor-immune interactions and a promising therapeutic target for enhancing immunotherapy efficacy. The mechanistic insights revealing how SOX9 orchestrates immunosuppression through multiple pathwaysâincluding the ANXA1-FPR1 axis in neutrophils, B7x checkpoint expression, and cancer stemness maintenanceâprovide a strong rationale for combinatorial approaches. Current preclinical data demonstrate that SOX9 inhibition can overcome resistance to combination checkpoint blockade, fundamentally reshaping the tumor immune microenvironment to favor cytotoxic T cell function and tumor control. Future research should prioritize the development of more specific and potent SOX9 inhibitors, validation across additional cancer types, and detailed exploration of timing and sequencing for optimal combination therapy. Translation of these findings to clinical application holds significant potential to expand the population of patients who benefit from immunotherapy and overcome key resistance mechanisms that currently limit long-term treatment success.
The accumulating evidence firmly establishes SOX9 as a critical node in cancer therapy resistance, particularly against modern immunotherapies. Its multifaceted role in promoting cancer stemness, epithelial-mesenchymal transition, and creating an immunosuppressive microenvironment positions SOX9 inhibition as a promising strategy to enhance treatment efficacy. Future research should prioritize the development of clinically viable SOX9 inhibitors, validate robust biomarkers for patient selection, and explore optimal sequencing and combination strategies with existing immunotherapies. Successfully targeting SOX9 represents a paradigm shift with the potential to overcome one of the most significant challenges in oncology todayâintrinsic and acquired therapy resistanceâultimately improving survival for patients across multiple cancer types.