Previous Article in Journal
Presence of Vaccine-Induced Antibodies Against Leptospira spp. Complicates the Diagnosis of Leptospirosis by the Microscopic Agglutination Test
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Therapeutic Strategies for Lung Cancer: The Role of Immunotherapy and HPV-Targeted Cancer Vaccines

School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
*
Author to whom correspondence should be addressed.
Vaccines 2025, 13(9), 957; https://doi.org/10.3390/vaccines13090957 (registering DOI)
Submission received: 30 July 2025 / Revised: 4 September 2025 / Accepted: 5 September 2025 / Published: 8 September 2025

Abstract

Background/Objectives: Lung cancer remains the leading cause of cancer-related deaths globally, with non-small-cell lung cancer (NSCLC) accounting for most cases. Although advances in targeted therapies and immunotherapy have improved outcomes, long-term survival remains limited. This review aims to explore current immunotherapeutic strategies, the evolving role of therapeutic cancer vaccines, and the emerging potential of human papillomavirus-targeted interventions in lung cancer, particularly among non-smoker populations. Methods: A comprehensive search of the literature was conducted using PubMed, Scopus, and Web of Science databases to identify relevant articles published between 2015 and 2024. Studies focusing on immune checkpoint inhibitors, vaccine platforms, HPV-associated lung cancer, tumor microenvironment modulation, and novel delivery systems such as bacterial ghosts were included. Relevant clinical trials and preclinical studies were critically evaluated and synthesized. Results: Immune checkpoint inhibitors targeting PD-1, PD-L1, and CTLA-4 have demonstrated clinical efficacy in NSCLC, yet their effectiveness is often limited by resistance mechanisms and lack of robust predictive biomarkers. Cancer vaccines, including peptide-based, mRNA, DNA, dendritic cell, and bacterial ghost platforms are emerging as complementary strategies to enhance antitumor immunity. Moreover, accumulating evidence suggests a potential association between high-risk HPV infection and lung cancer development, supporting the rationale for HPV-targeted vaccine strategies. Conclusions: Immunotherapy and therapeutic vaccination hold significant promise in reshaping lung cancer treatment. Advancements in vaccine design, delivery platforms like bacterial ghosts, and better understanding of HPV’s role in lung oncogenesis could support more effective, personalized immunotherapeutic approaches in the future.

1. Introduction

Lung cancer is the most diagnosed and deadliest cancer worldwide. According to GLOBOCAN (a cancer observatory maintained by the International Agency for Research on Cancer (IARC), which compiles and standardizes international cancer incidence, prevalence, and mortality data) 2020, there were approximately 2.2 million new lung cancer cases (11.4%) and 1.8 million deaths (18%) reported worldwide [1]. The distribution and impact of lung cancer differ greatly across regions. This variation is influenced by factors like smoking rates, environmental pollution, work-related dangers, and diet. In the United States, the lung cancer rate has steadily declined since 1990. This decrease is mainly due to effective tobacco control policies [2]. Despite these improvements, lung cancer remains a major public health issue globally. Figure 1 indicates worldwide cancer cases, where lung cancer remains among the top contributors to the global cancer burden, emphasizing the critical need for improved therapeutic strategies.
Traditional lung cancer treatments, such as surgery, radiation therapy, chemotherapy, and targeted drug therapies, have improved over time [3]. However, these treatments often face challenges like high relapse rates, the development of drug resistance, and significant side effects. Specifically, chemotherapy and radiation therapy, whether used before or after surgery, provide only modest improvements in survival and are often linked with toxic effects. As a result, the prognosis for metastatic lung cancer is still poor; five-year survival rates are around 4% [4]. Smoking causes more than 80% of lung cancer deaths, but other major risk factors include radon exposure, asbestos, long-term contact with air pollutants like polycyclic aromatic hydrocarbons (PAHs), and a family history of lung cancer [5]. The World Health Organization divides lung cancers mainly into two categories: small-cell lung cancer (SCLC), which makes up about 15% of cases, and non-small-cell lung cancer (NSCLC), accounting for approximately 80–85% of cases. NSCLC can be further divided into adenocarcinoma (LUAD), squamous-cell carcinoma (LUSC), and large-cell carcinoma (LCC), each with distinct molecular and genetic profiles. Lung cancer is a significant focus in cancer immunology research due to frequent mutations in proto-oncogenes and tumor suppressor genes, as well as issues with the immune system [3]. The limitations of conventional therapies have sparked growing interest in more personalized, immune-based treatment options. Currently, low-dose computed tomography (LDCT) is the best method for early lung cancer screening, but preventing and treating the disease remains difficult. This has led to a rise in research on immunotherapy and therapeutic cancer vaccines as promising new approaches [6]. Immunotherapy seeks to use or adjust the immune system to target tumor cells selectively, often focusing on tumor-specific antigens.
A noteworthy area of interest is the possible role of human papillomavirus (HPV) in lung cancer development, especially among non-smokers. HPV is mainly known for infecting mucosal tissues in the mouth, throat, and genital areas. HPV is primarily known for its causal role in cervical cancer, where the viral oncoproteins E6 and E7 inactivate key tumor suppressors such as p53 and Rb. Similar mechanisms are well documented in the development of anogenital and head-and-neck cancers, and these parallels suggest that HPV-driven molecular pathways may also contribute to lung carcinogenesis. Researchers suggest it may play a role in lung cancer, although its exact contribution is still being studied [7]. Other viruses linked to lung cancer include retroviruses, Epstein–Barr virus, and human immunodeficiency virus. However, HPV is notable for its presence in about 31% of lung cancer cases [8]. Advanced molecular biology techniques have shown higher HPV rates in malignant lung tissues compared to benign or normal samples. High-risk HPV strains, especially HPV16 and HPV18, along with their viral oncoproteins E6 and E7, are thought to be key in the cancer process [9]. This review aims to examine new treatment strategies for lung cancer, focusing on immunotherapy and HPV-targeted cancer vaccines. It will look closely at the shortcomings of current treatments, highlight recent advancements in immunological approaches, and evaluate evidence about HPV’s role in lung cancer development. By integrating current findings, the review seeks to identify promising targeted therapies that might improve clinical outcomes and guide future research efforts.

2. Epidemiology and Risk Factors of Lung Cancer

Lung cancer is the one of the most diagnosed cancers worldwide and the leading cause of cancer-related death. It accounted for about 13% of all new cancer cases in 2020 [1]. It is the most common cancer among men and the second most common among women, with around 2.2 million new cases worldwide that year. Additionally, lung cancer caused about 1.8 million deaths in 2020, highlighting its heavy impact on global public health [10]. Differences in lung cancer rates show how smoking habits and tobacco control measures vary by region. While incidence has declined in high-income countries due to reduced smoking, rates are rising in low- and middle-income regions, particularly in the Asia-Pacific, where non-smoking-related cases are increasingly reported [11]. These variations mainly result from historical tobacco-use patterns and the effectiveness of public health policies aimed at reducing smoking [12]. Research shows that strong nicotine control programs can lower lung cancer deaths by up to 40%, emphasizing the importance of ongoing tobacco control efforts [13].
Tobacco smoking has long been known as the main risk factor, accounting for over 80% of lung cancer cases each year [14]. Recent research stresses the need to consider all related risk factors without bias. Smokers face a 10- to 15-fold higher risk of developing lung cancer compared to non-smokers, with about 85% of cases linked to active smoking [15]. The risk is higher not only for current smokers but also for former smokers, people exposed to second-hand smoke, and users of other tobacco products [12]. The harmful substances in tobacco smoke cause destructive, growth-promoting, and transforming changes in bronchial cells, leading to cancer via genetic and epigenetic changes [16]. Studies have shown that these carcinogens harm various oncogenes and tumor suppressor genes, especially in lung tissue [17]. For non-smokers, second-hand smoke is the main risk factor, highlighting the need for effective smoke-free policies [18]. In addition to tobacco, air pollution has become a major environmental risk factor for lung cancer. Elderly people living in urban areas with high levels of fine particulate matter (PM2.5) face increased risk [19]. Research shows a dose–response relationship between PM2.5 exposure and lung cancer rates, with some studies finding increased risk even below current regulatory limits [20].
Occupational and environmental exposures also significantly raise lung cancer risk. For example, exposure to asbestos alone increases the risk of developing lung cancer by about 3 to 10 times, a risk that rises dramatically when combined with cigarette smoking—over a 50-fold increase in some studies [21,22]. Other workplace hazards linked to higher lung cancer risk include silica dust, arsenic, and certain industrial chemicals [23]. Beyond chemical and particulate exposures, viral infections are an emerging area of interest in lung cancer etiology. Among these, human papillomavirus (HPV) has gained attention, particularly in non-smoking populations. HPV, a DNA virus known for its role in anogenital and oropharyngeal cancers, has also been implicated in pulmonary carcinogenesis. High-risk HPV subtypes—especially HPV16 and HPV18—have been identified in malignant lung tissues, and their E6 and E7 oncoproteins are believed to disrupt p53 and Rb tumor suppressor pathways, promoting uncontrolled cell proliferation [8,9]. Multiple molecular epidemiological studies have reported HPV DNA in lung tumor samples, with prevalence estimates ranging from 20% to over 30% depending on geographic and population-specific factors [7]. A meta-analysis further supports the association between HPV and lung cancer, particularly among non-smokers and women. Moreover, the use of advanced detection techniques such as polymerase chain reaction (PCR) has improved the sensitivity of HPV identification in lung tissue samples, lending further credibility to these findings [24]. Although the causal role of HPV in lung cancer remains under investigation, the observed correlations suggest a potentially important etiological factor, especially in regions with high HPV prevalence and among patient populations without significant tobacco exposure. Recognition of HPV’s oncogenic potential in lung cancer may open new avenues for prevention, including targeted vaccination and immunotherapy strategies.

Controversies and Limitations of Current Evidence

Although multiple studies have reported a significant association between high-risk HPV infection and lung cancer, particularly in non-smokers, it remains unclear whether HPV plays a causative role. Detection of HPV DNA or proteins in tumor samples may reflect correlation rather than direct oncogenic involvement. Confounding factors, such as geographic differences, sample contamination, and methodological variability, complicate interpretation. Furthermore, the relatively low prevalence of HPV-positive lung cancers compared to cervical and head-and-neck cancers suggests that HPV may act as a co-factor rather than a primary driver. Future studies employing standardized assays and large, multicenter cohorts are required to clarify whether HPV is causally involved in lung carcinogenesis or merely associated with disease in certain populations.

3. Molecular Classification and Pathogenesis

Lung cancer falls into two main categories: non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC). Each category has different molecular and clinical profiles. NSCLC makes up about 85% of lung cancers and has three main subtypes: lung adenocarcinoma (LUAD), lung squamous-cell carcinoma (LUSC), and large-cell carcinoma (LCC). SCLC accounts for the remaining 15%. It is known for rapid progression, high potential to spread, and neuroendocrine characteristics [25]. Lung cancer arises from diverse molecular alterations, including mutations in KRAS, EGFR, TP53, and chromosomal rearrangements such as ALK fusions. Beyond genetic drivers, viruses are established contributors to carcinogenesis, as seen with hepatitis B virus in hepatocellular carcinoma, Epstein–Barr virus in nasopharyngeal carcinoma, and human papillomavirus in cervical and anogenital cancers. Within this broader context, HPV has emerged as a causative candidate in lung cancer [26,27].
High-risk human papillomavirus (HPV) types, particularly HPV-16 and HPV-18, have been implicated in lung carcinogenesis through the expression of oncoproteins E6 and E7. These proteins inactivate key tumor suppressors such as p53 and Rb, promoting uncontrolled cell proliferation and genomic instability. Figure 2 illustrates the proposed mechanism by which HPV enters the lung epithelium, disrupts cellular regulatory pathways, and contributes to tumor progression and metastasis.

3.1. SCLC vs. NSCLC Subtypes (LUAD, LUSC, LCC)

LUAD usually develops in the outer regions of the lung and is the most common type among non-smokers. It is often linked to mutations in EGFR, KRAS, ALK, BRAF, and MET. LUSC is found in the center of the lung and is strongly associated with smoking; it shows changes in TP53, PIK3CA, CDKN2A, and SOX2. LCC is a diagnosis made when other types are excluded; it represents undifferentiated tumors lacking glandular or squamous features. This type has a high mutation rate but few mutations that can be targeted [28]. SCLC is marked by the loss of function in both TP53 and RB1. It often shows neuroendocrine markers like ASCL1 and NEUROD1. Molecular profiling of SCLC has led to four subtypes based on the expression of transcription factors: ASCL1-high (SCLC-A), NEUROD1-high (SCLC-N), POU2F3-high (SCLC-P), and an inflammatory subtype (SCLC-I). The inflammatory subtype may have a better response to immunotherapy [29,30].

3.2. Genetic Mutations and Immune Evasion Mechanisms

The mutation patterns in LUAD and LUSC greatly affect tumor biology and treatment options. Immune exclusion refers to a tumor immune evasion strategy where immune cells, particularly cytotoxic T lymphocytes, are physically prevented from infiltrating the tumor parenchyma, often due to stromal barriers, abnormal vasculature, or immunosuppressive cells within the tumor microenvironment. In LUAD, EGFR mutations are linked to immune exclusion and a lower tumor mutation burden (TMB), which leads to a reduced response to immune checkpoint inhibitors (ICIs) [31]. On the other hand, KRAS-mutant LUAD, especially with concurrent TP53 mutations, often shows high PD-L1 expression and immune cell infiltration. This can enhance responsiveness to ICIs [32]. Immune evasion is a key feature of lung cancer. Tumors reduce the expression of MHC class I molecules. They secrete immunosuppressive cytokines, like TGF-β and IL-10, and recruit regulatory T cells and myeloid-derived suppressor cells (MDSCs) to limit the activity of cytotoxic T cells [33].

4. Immunotherapy in Lung Cancer

Lung cancer treatment relies on a combination of traditional and emerging modalities, including surgery, chemotherapy, radiotherapy, targeted therapy, immunotherapy, and cancer vaccines. As shown in Figure 3, these treatment strategies vary depending on cancer stage, molecular profile, and patient-specific factors.
Immunotherapy has greatly changed the treatment options for lung cancer, especially non-small-cell lung cancer (NSCLC). It offers better survival rates and lasting responses in certain patient groups. Among the different immunotherapy approaches, immune checkpoint inhibitors (ICIs) that target programmed cell death protein-1 (PD-1), its ligand PD-L1, cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and the recently studied lymphocyte-activation gene 3 (LAG-3) have shown significant effectiveness [34,35]. These drugs work by blocking immune-suppressive pathways used by tumors to hide from the immune system, thus restoring T cell activity against tumors. T cell activation is carefully regulated by a complex interaction of co-stimulatory and co-inhibitory signals. Immune checkpoints help manage this balance to prevent excessive immune reactions and autoimmunity. However, in cancer, tumor cells often manipulate these checkpoints to weaken T cell function [36]. Therapeutic antibodies that block PD-1/PD-L1 and CTLA-4 pathways enable T cell growth and cytokine production, making tumors more recognizable to the immune system. Nivolumab and pembrolizumab, which target PD-1, are now approved for advanced NSCLC and have shown lasting clinical responses in certain patients [37]. Immunotherapy has emerged as a cornerstone in lung cancer treatment, particularly through immune checkpoint blockade targeting PD-1/PD-L1 and CTLA-4 pathways. These therapies restore the ability of T cells to recognize and eliminate tumor cells. Figure 4 illustrates the mechanisms by which checkpoint inhibitors modulate the tumor–immune interaction within the lung tumor microenvironment.
In addition to checkpoint blockers, other immunotherapy methods are gaining popularity. These include chimeric antigen receptor T cell (CAR-T) therapy, cytokine-induced killer (CIK) cells, and combination therapies that use different immune-boosting agents together. CAR-T cell therapy, while previously studied mainly in blood cancers, is being explored for solid tumors like lung cancer. However, challenges such as suppression in the tumor microenvironment (TME) and differences in tumor antigens remain [38]. Combining immunotherapy with other treatments, such as chemotherapy or antiangiogenic therapies, has shown promise in increasing immune responses and reducing resistance [39]. The tumor microenvironment (TME) significantly affects immune responses. Elements within the TME, such as regulatory T cells, myeloid-derived suppressor cells, and immunosuppressive cytokines, create an environment that makes effective immunotherapy difficult. Addressing these challenges is crucial for enhancing treatment responses and is a key focus of current research [40]. Despite positive results, there are still challenges. One major issue is the inconsistent predictive value of PD-L1 expression in both NSCLC and small-cell lung cancer (SCLC). While PD-L1 is often more abundant in immune cells infiltrating tumors than in the tumors themselves, it has not reliably predicted clinical benefit in SCLC, which limits its usefulness as a biomarker [41]. As a result, researchers are looking for alternative targets. Molecules like B7-H3 and CD47 are emerging as promising options. B7-H3 is believed to limit T cell entry and support tumor growth, while CD47 serves as a “don’t eat me” signal that stops the immune system from attacking tumor cells. Blocking CD47 has shown promise in improving the removal of SCLC cells by the immune system [42].
Immunotherapy is a groundbreaking approach in treating lung cancer, with ICIs being a central part of current clinical practices. Although immune checkpoint inhibitors have transformed lung cancer treatment, they are associated with immune-related adverse events (irAEs), including pneumonitis, colitis, dermatitis, and endocrinopathies. These off-target toxicities distinguish immunotherapy from chemotherapy and targeted therapies, highlighting the importance of biomarkers that can predict not only treatment response but also toxicity risk. Table 1 summarizes the challenges in immunotherapy and their proposed strategies with ongoing clinical trials. Developing new targets, optimizing combination therapies, and gaining a better understanding of immune evasion and TME will be vital for maximizing effectiveness and expanding the advantages of immunotherapy to more patients.

5. Cancer Vaccines in Lung Cancer

5.1. Traditional and Neoantigen-Based Vaccines

Cancer vaccines have traditionally targeted infectious diseases. Recently, they were being adapted to stimulate immune responses against cancer. Early therapeutic cancer vaccines focused on tumor-associated antigens (TAAs), which are self-proteins overexpressed in tumors. However, these vaccines often failed to trigger effective new T cell responses because of immune tolerance mechanisms [47]. In contrast, neoantigen-based vaccines target tumor-specific antigens (TSAs) that come from somatic mutations and are not affected by central or peripheral tolerance. They provide a strong way to trigger targeted immune responses with limited off-target effects. These vaccines also tend to avoid unwanted damage to normal tissues [48,49]. Finding and selecting appropriate neoantigens that are uniquely expressed by cancer cells is critical for vaccine effectiveness. Unlike TAAs, neoantigens come from mutated proteins that the immune system recognizes as foreign, usually leading to strong CD4⁺ and CD8⁺ T cell responses [49]. However, tumors often contain numerous mutations, which increases the chances of finding immunogenic neoantigens in cancers with high mutational burdens. Even with careful target selection, the immunosuppressive tumor microenvironment (TME) poses a significant challenge. This environment is marked by low oxygen, lack of nutrients, acidic pH, high levels of reactive oxygen species, and dense infiltration by regulatory T cells [50].
In addition to TME challenges, several other factors limit vaccine performance. Tumor fibroblasts, myeloid-derived suppressor cells, and a thick extracellular matrix hinder T cell infiltration [50]. Low levels of antigen-presenting cells (APCs), mechanisms for antigen escape, and the overall tumor mutational burden also restrict vaccine responses. Although neo-antigen strategies have made great strides, many remain in preclinical stages. Vaccination alone may not be enough without addressing the broader immunosuppressive context. It is important to combine adjuvants, proper antigen selection, and effective delivery systems to overcome these barriers. Currently, the most advanced vaccine platforms include cellular vaccines, which can be based on whole tumor cells or dendritic cells (DC), synthetic long-peptide (SLP) vaccines, and nucleic acid-based vaccines (RNA or DNA) [51]. Table 2 summarizes different vaccine platforms for lung cancer in the developmental stage.

5.2. Nucleic Acid Vaccines

Nucleic acid vaccines use mRNA or DNA that encode tumor antigens, which host cells take up and translate in the body. mRNA vaccines allow for quick development and strong immune response, making them ideal for personalized neoantigen strategies or general use [60]. Crucially, mRNA allows for post-translational modifications like glycosylation and phosphorylation, which are often essential for proper protein folding and antigen presentation. Directly injecting mRNA into lymph nodes has been shown to activate APCs, marked by increased CD86 and IL-12 expression, and stimulate strong CD4⁺ and CD8⁺ T cell responses [61]. DNA vaccines have advantages in terms of stability and storage. Plasmid DNA can produce multiple mRNA copies once transcribed in the nucleus. However, this requirement for entering the nucleus delays the production of antigens and lowers their immune response compared to mRNA vaccines. DNA also carries a potential, though low, risk of integrating into the genome, a risk that mRNA vaccines do not have [40]. Mechanically, DNA vaccines involve inserting plasmids encoding tumor antigens into somatic cells, leading to antigen expression and presentation through major histocompatibility complex (MHC) I and II pathways [62]. Antigens can be expressed inside cells, secreted, or released in apoptotic bodies, stimulating both humoral and cellular immunity. The best delivery method seems to be direct transfection of APCs, especially through intradermal injection [63]. DNA vaccines also activate innate immune pathways—through TLR9 signaling and STING activation—resulting in the release of chemokines, cytokines, and type I interferons [64]. These vaccines are cost-effective, stable, safe, and specific, yet their moderate immune response has limited their success in clinical settings so far [65,66].

5.3. Peptide-Based Cancer Vaccines

Peptide vaccines generally consist of 20–30 amino acids that represent TAAs or TSAs, including neoantigens or cancer/testis antigens. These peptides are designed to trigger both CD8⁺ and CD4⁺ T cell responses via MHC class I and II presentation [67]. Synthetic long peptides (SLPs) are preferred because their processing inside cells leads to stronger T cell activation compared to short peptides [68]. To boost immune responses, peptide vaccines are formulated with adjuvants like aluminum salts, MF59, CpG motifs, poly-ICLC, and cyclic dinucleotides. Heteroclitic peptides, modified to enhance MHC binding, further increase immune response. Because peptides can degrade quickly in the body, they are often delivered using stabilizing carriers like liposomes or PLGA nanoparticles [69]. To tackle the immunosuppressive TME, future cancer vaccine strategies should continue to optimize antigen selection, delivery methods, and adjuvant systems. By combining strong neoantigen identification with advanced vaccine design, we can enhance the chances of generating lasting, specific immune responses in lung cancer patients.

5.4. Bacterial Ghost-Based Cancer Vaccine

Bacterial ghosts (BGs) are empty bacterial cell envelopes generated by genetic engineering or chemical methods. They maintain the native surface antigens and structural integrity of bacteria but are devoid of cytoplasmic contents, making them a promising platform for safe and immunogenic antigen delivery. BGs can be engineered to carry tumor-associated antigens or adjuvants and have shown efficacy in preclinical models for inducing strong Th1-skewed immune responses, activating antigen-presenting cells, and promoting cytotoxic T cell activation. Given their intrinsic adjuvanticity and ability to mimic pathogen-associated molecular patterns (PAMPs), BG-based vaccines are particularly promising for lung cancer immunotherapy. While research is still emerging, recent studies have explored their application in mucosal immunization and intratumoral delivery in lung and colorectal cancer models. Many probiotic bacteria are also used for bacteria-based cancer vaccines. Their use could complement existing cancer vaccine strategies by enhancing innate and adaptive immune activation with minimal systemic toxicity [53,70].
Peptide vaccines, killed virus, and BGs primarily promote antigen-specific CD4⁺ T helper responses and CD8⁺ expansion with cytokine secretion. True cytotoxic activity requires endogenous antigen synthesis (e.g., live virus, mRNA, DNA vaccines). Therefore, BGs and peptide vaccines serve more as immune modulators than direct inducers of cytotoxic clearance. While bacterial ghosts and peptide vaccines can induce CD8⁺ T cell expansion and IFN-γ secretion, these responses may not translate into direct cytotoxic activity against tumor cells unless combined with antigen delivery strategies that support endogenous processing.

6. Vaccine Adjuvants for Enhancing Cancer Immunity

The effectiveness of cancer vaccines relies not just on choosing tumor-specific antigens but also on having suitable adjuvants that can enhance and guide the immune response. Adjuvants improve the immunogenicity of vaccines by stimulating antigen-presenting cells (APCs), promoting antigen uptake, and initiating both innate and adaptive immunity. This section categorizes vaccine adjuvants into traditional and emerging classes, summarizing how they work and their clinical relevance in cancer immunotherapy.

6.1. Traditional Adjuvants: TLR Agonists, Aluminum Salts, Poly-ICLC

Conventional adjuvants have formed the basis for many immunotherapies. Among the most used are toll-like receptor (TLR) agonists, aluminum-based compounds, and synthetic double-stranded RNA analogs like polyinosinic–polycytidylic acid stabilized with polylysine and carboxymethylcellulose (poly-ICLC). These substances mainly work by activating pattern recognition receptors (PRRs), signaling the immune system to respond to perceived threats [71]. TLR agonists, like CpG oligodeoxynucleotides (TLR9), imiquimod (TLR7), and monophosphoryl lipid A (TLR4), have shown strong immunostimulatory effects by improving dendritic cell (DC) maturation and boosting type I interferon secretion [72]. Poly-ICLC, which mimics viral RNA, works through TLR3 and the melanoma differentiation-associated protein 5 (MDA5) pathway to stimulate cytokine production and T cell activation. Aluminum salts, while traditionally used in preventive vaccines, are less effective in generating the cytotoxic T lymphocyte (CTL) responses needed for tumor rejection, but they may still support humoral immunity through inflammasome activation. While adjuvants such as alum are effective in enhancing humoral responses, their role in promoting T cell responses in tissue-resident settings remains controversial, with some formulations raising concerns of excessive inflammation or tissue adhesion, particularly in the context of HPV vaccines.

6.2. Emerging Adjuvants: STING Agonists, CD40, Cytokines, and Inorganic Nanoparticles

Recent studies have discovered a new generation of adjuvants that could overcome the immunosuppressive tumor microenvironment (TME) and enhance antigen presentation. These include STING agonists, CD40 agonists, immune-boosting cytokines (e.g., GM-CSF), and engineered inorganic nanoparticles.
STING (stimulator of interferon genes) agonists are a promising group of adjuvants that activate strong type I interferon responses through cytosolic DNA sensing pathways. Natural cyclic dinucleotides (CDNs) and synthetic derivatives such as cyclic GMP–AMP (cGAMP) have shown success in activating antitumor immunity through STING-mediated signaling [73,74,75]. These agents enhance DC maturation and CD8+ T cell priming and increase the infiltration of effector T cells into tumors.
CD40 agonists stimulate DCs and B cells, improving antigen processing and cross-presentation. Although they were initially studied as standalone treatments, CD40 ligands have shown better results when combined with TLR agonists in preclinical vaccine models [67]. However, their clinical use has been limited due to potential toxicity and variable pharmacokinetics. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is one of the most studied cytokine adjuvants. It promotes DC recruitment and activation at the vaccination site [76]. Despite positive preclinical results, clinical trials have had mixed outcomes. GM-CSF can lead to an increase in immunosuppressive myeloid-derived suppressor cells (MDSCs) under certain conditions, which may counteract its intended effects [77,78,79]. These conflicting effects highlight the need for better dosing strategies and combination treatments.
Inorganic nanoparticles, such as titanium dioxide (TiO2) and manganese-based adjuvants, have emerged as innovative delivery systems and immune boosters. For example, spiky TiO2 nanoparticles disrupt phagolysosomal membranes during APC uptake, improving antigen cross-presentation and T cell priming [80,81]. Manganese can enhance STING signaling and cytosolic DNA sensing. Manganese-based colloidal gels (e.g., MJ) significantly improved both humoral and cellular immune responses in preclinical cancer models [82]. Other experimental platforms include polymeric nanoparticles (e.g., PC7A) that combine antigen delivery with STING activation and better T cell responses [83,84]. Additionally, compounds that modify the mevalonate pathway—like statins and bisphosphonates—are being studied for their ability to generate strong CTL responses by changing cholesterol biosynthesis and improving antigen presentation [85].

6.3. Clinical and Preclinical Findings

Many adjuvants have shown promise in preclinical models, but translating these results into human use remains difficult. Challenges include concerns about toxicity, differences in immune responses, and limited information on long-term safety. GM-CSF is still used in clinical trials, despite its mixed results [86]. New platforms like STING and CD40 agonists have shown potential in early studies, but they need careful optimization to prevent systemic inflammation. Similarly, controlling the physicochemical properties of nanoparticle-based adjuvants is essential to balance effectiveness with compatibility. In the end, the ideal cancer vaccine adjuvant should safely and effectively boost tumor-specific immunity, help with antigen presentation, and modulate the tumor microenvironment without causing significant off-target effects. Thoughtful design, combination strategies, and personalized approaches based on biomarkers will be crucial for the next generation of adjuvants for lung cancer vaccines.

7. Imidazoquinolines (IMDs) as Cancer Immunomodulators

7.1. Mechanisms: TLR7/8 Activation

Imidazoquinolines (IMDs) are a class of synthetic small-molecule agonists that activate toll-like receptors 7 and 8 (TLR7/8), which are localized in endosomal compartments of innate immune cells. Upon activation, these receptors trigger a MyD88-dependent signaling cascade involving IRAK1/4 and TRAF6, culminating in the activation of NF-κB and mitogen-activated protein kinases (MAPKs), as well as interferon regulatory factors (IRFs) [87,88]. This cascade stimulates the production of proinflammatory cytokines such as IL-12 and type I interferons (IFNs), particularly IFN-α, which enhance antigen presentation, promote dendritic cell (DC) maturation, and improve cytotoxic T lymphocyte (CTL) activation [89]. These mechanisms have established IMDs as potent immunomodulators in oncology. However, the biological effects of TLR7/8 ligands can vary depending on tumor type and microenvironment. For instance, while some preclinical studies demonstrated tumoricidal activity through enhanced adaptive immunity, others have shown that TLR7/8 activation might paradoxically promote lung tumor progression in certain contexts [90,91]. These dichotomous findings underscore the necessity of precise targeting strategies and context-aware therapeutic design [92,93].

7.2. FDA-Approved Agents: Imiquimod and Resiquimod

The first FDA-approved IMD was imiquimod (R837), licensed in 1997 for the topical treatment of genital and perianal warts, actinic keratosis, and later, superficial basal-cell carcinoma [94]. It selectively activates TLR7, inducing robust local immune responses through type I IFN production. Building upon this success, resiquimod (R848) was developed as a more potent TLR7/8 dual agonist—estimated to be over 100 times more effective in TLR activation than R837 [95]. While R848 has not yet received FDA approval, it has shown substantial immunostimulatory capacity in preclinical models and early-phase clinical trials. Systemic administration of R848 in patients with metastatic solid tumors (e.g., colorectal, breast, ovarian, and cervical cancers) led to disease stabilization in a subset of cases and elevated serum levels of IFN-γ and TNF-α, indicative of heightened immune activation [96]. However, systemic cytokine release also induced significant adverse effects, limiting further clinical progression. These challenges have redirected focus toward topical or targeted delivery methods for safer, localized immunomodulation [95].

7.3. Applications as Vaccine Adjuvants and Topical Immunotherapies

IMDs such as imiquimod and resiquimod are increasingly used as vaccine adjuvants, particularly in therapeutic cancer vaccines. More than 100 clinical trials have explored their role as immune potentiators in vaccines for cancer (e.g., dendritic cell-based immunizations), infectious diseases (e.g., hepatitis B), and respiratory illnesses (e.g., influenza) [97]. Recent studies suggest that combining IMDs with tumor-associated or viral antigens—such as HPV16 E7—may offer a viable immunotherapeutic strategy for HPV-associated lung cancers. Topical IMD formulations are of particular interest due to their ability to generate robust local inflammation and antigen-specific immune responses without systemic toxicity. These benefits are driven by TLR7/8-mediated cytokine release, particularly IL-12 and type I IFNs, which activate plasmacytoid DCs, conventional DCs, macrophages, and monocytes [95,98]. These mechanisms support their dual utility in both topical immunotherapy and vaccine adjuvancy.

7.4. Future Directions and Clinical Integration

Despite their promising effects on the immune system, the use of IMDs in the whole body is still limited due to the risk of cytokine storms and other inflammatory reactions. Future strategies focus on localized delivery systems, like nanoformulations or injections directly into tumors, to improve effectiveness while reducing side effects [96]. Table 3 illustrates the mechanism of action and clinical status of traditional and emerging adjuvants. Research is also looking into changing the structure of IMDs to adjust TLR specificity and improve signaling strength [95].

8. Future Perspectives and Research Directions

8.1. Personalized Neoantigen Vaccines and Immunotherapy

Personalized cancer immunotherapy, especially through neoantigen-based vaccination strategies, has quickly moved from concept to early clinical practice. These methods, particularly when combined with immune checkpoint inhibitors, are creating new possibilities for cancer treatment. As this area develops, future research will likely focus on refining and broadening four key areas: personalized vaccine strategies, combinations of different immunotherapies, integrating virology with oncology, and developing predictive biomarkers to guide treatment. Table 4 represents the feasibility and timeline of immunotherapeutic strategies.
Personalized cancer vaccines are designed to take advantage of tumor diversity by targeting patient-specific neoantigens that arise from non-synonymous mutations [99]. Unlike shared tumor-associated antigens, neoantigens are not found in healthy tissues. This significantly lowers the risk of central immune tolerance and autoimmune reactions. Several types of vaccine platforms have been tested, including synthetic long peptides, mRNA, DNA, dendritic cell-based vaccines, and viral vector vaccines. Currently, mRNA platforms lead due to their flexibility, rapid production, and safety [100].
Clinical data show that neoantigen vaccines are feasible and can provoke immune responses across multiple tumor types. For example, the KEYNOTE-942 trial showed that the mRNA-4157 vaccine from Moderna and Merck, used alongside pembrolizumab, led to a 44% decrease in melanoma recurrence compared to checkpoint blockade used alone [101]. Similarly, trials in glioblastoma and non-small-cell lung cancer have found that personalized peptide vaccines can generate lasting CD4+ and CD8+ T cell responses when combined with standard treatments [102].
Future work on vaccine strategies will involve improving neoantigen prediction algorithms by using multiomics data and machine learning. Additionally, researchers will optimize delivery methods, such as lipid nanoparticles and viral vectors, while working to lower the time and cost of making individualized vaccines. Progress in RNA technology, including self-amplifying mRNA and circular RNA platforms, could enhance antigen expression and reduce dosage needs, which may improve effectiveness and accessibility [103].

8.2. Multi-Modal Immunotherapy Combinations

A promising future direction is combining neoantigen vaccines with other immunotherapies. Checkpoint inhibitors, like anti-PD-1 and anti-CTLA-4 antibodies, help T cell function by preventing exhaustion. When used alongside personalized vaccines, these agents show synergistic effects. Vaccines expand the antigen-specific T cell pool while checkpoint inhibitors sustain their activity [104].
In addition to checkpoint inhibitors, combining neoantigen vaccines with adoptive T cell therapies, such as chimeric antigen receptor (CAR) T cells or tumor-infiltrating lymphocytes (TILs), is gaining interest. Vaccine priming might improve the efficacy of infused T cells by broadening the pool of relevant antigen-specific clones. Oncolytic viruses, which specifically target tumor cells, can increase local inflammation and antigen presentation, acting as additional vaccine boosters [105].
Timing will be crucial for the success of these combinations. Research suggests that administering vaccines before or at the same time as checkpoint inhibitors may maximize effectiveness. A delayed administration of checkpoint inhibitors could reduce their synergistic effect due to early T cell exhaustion [106]. Ongoing clinical trials are exploring the best schedules and dosing for these combinations.

8.3. Integrating Virology and Oncology

Another emerging focus is to combine virology with personalized immunotherapy. Virus-related cancers, such as cervical cancer (HPV), nasopharyngeal carcinoma (EBV), and Merkel cell carcinoma (polyomavirus), provide additional targets through viral oncoproteins. These viral antigens make ideal immunologic targets, since they are foreign and highly conserved, allowing for both personalized and semi-universal vaccine designs [107].
Moreover, researchers are considering viral vectors as delivery vehicles for neoantigen vaccines. Their strong ability to provoke immune responses and infect antigen-presenting cells is beneficial [105]. Modified adenoviruses, poxviruses, and lentiviruses are being tested in both lab and clinical settings. Oncolytic viruses may offer two benefits: they can directly destroy tumors and stimulate the release of immunogenic antigens, enhancing the effectiveness of personalized vaccines when used together. A future strategy might involve engineering synthetic viruses that carry both neoantigens and immunomodulatory genes like GM-CSF or IL-12. This vitro-immunotherapeutic approach aims to boost both innate and adaptive immune responses while reducing systemic toxicity [100].

8.4. Predictive Biomarkers and Precision Approaches

A vital future focus is identifying and validating biomarkers that predict which patients will benefit the most from neoantigen vaccine therapies. Tumor mutational burden (TMB) has often served as a measure of neoantigen load, but it varies across different tumor types and individual patients [106]. Not all mutations create immunogenic peptides, and some tumors with high TMB still escape immune detection due to faulty antigen processing.
Other potential biomarkers include the clonality of neoantigens, MHC-binding affinity scores, CD8+ T cell density in tumors, PD-L1 expression, and IFN-γ gene signatures [108]. Circulating tumor DNA (ctDNA) is increasingly used to track treatment responses in real time. For instance, drops in ctDNA levels after vaccination are linked to better progression-free survival in melanoma and glioblastoma [100].
Advances in spatial transcriptomics and multiplexed immunohistochemistry allow researchers to map immune cells and neo-antigen expression within the tumor microenvironment. These technologies can help customize vaccine design by selecting antigens from regions rich in immune cells while steering clear of areas lacking immune activity [105].
While cancer vaccines have focused largely on host-derived neoantigens and tumor-associated antigens, HPV represents a distinct viral target in a subset of lung cancers. HPV-targeted vaccines remain largely unexplored for lung cancer, but preclinical studies suggest that vaccines targeting HPV E6 and E7 oncoproteins may enhance antitumor immunity in HPV-positive lung tumors. Translation to the clinic is limited by the relatively low prevalence of HPV-positive lung cancers and the lack of standardized diagnostic assays. Nevertheless, lessons from successful HPV-targeted vaccines in cervical and head-and-neck cancers provide a foundation for developing lung cancer vaccines aimed at viral causes. Future strategies may involve combination regimens that integrate HPV vaccines with checkpoint inhibitors or novel adjuvants to increase efficacy.
Ultimately, the aim is to create integrated biomarker models combining genomic, proteomic, and immunologic data using machine learning algorithms. These models will assist in choosing patients, formulating vaccines, and planning combination strategies. They will enable real-time adjustments to immunotherapy regimens based on changes in tumors, creating a truly dynamic precision oncology environment.
The future of cancer immunotherapy is in merging personalized neoantigen vaccines with multi-modal strategies, viral immunology, and precise biomarker frameworks. As clinical experiences increase and technology improves, we expect better access, enhanced outcomes, and adaptive treatment models to emerge. By focusing on refining antigen prediction, validating combination therapies, and expanding biomarker-guided strategies, the field is set to provide effective immunotherapy to more patients.

9. Conclusions

Lung cancer continues to be one of the most challenging malignancies to treat, largely due to its late-stage diagnosis, molecular heterogeneity, and complex immune evasion mechanisms. The advent of immunotherapy, particularly immune checkpoint inhibitors, has revolutionized the therapeutic landscape and significantly improved outcomes for a subset of patients. However, treatment resistance, limited biomarker precision, and variability in response highlight the need for more refined and innovative approaches.
Therapeutic cancer vaccines represent a promising adjunct to existing immunotherapies, aiming to generate durable and antigen-specific immune responses. Among these, platforms such as mRNA, DNA, dendritic cell, and bacterial ghost-based vaccines are under active investigation, each offering unique immunogenic and logistical advantages. Notably, emerging evidence implicates high-risk HPV in the pathogenesis of lung cancer, particularly among non-smoking individuals, suggesting a potential new avenue for HPV-targeted therapeutic strategies.
Future research must focus on optimizing vaccine delivery, identifying reliable predictive biomarkers, and integrating immunotherapy with conventional and targeted treatments. Platforms like bacterial ghosts also present an exciting frontier, offering both antigen presentation and innate immune activation in a single system. While BGs represent a novel avenue for vaccine delivery and immunomodulation, their safety and efficacy should be validated in preclinical and clinical settings. Moreover, reliable biomarkers are essential for predicting response and toxicity to immunotherapies, enabling more precise patient selection in biomarker discovery. Overall, the integration of immunotherapy, vaccine science, and virology holds the key to more personalized, effective, and long-lasting treatments for lung cancer. Continued exploration and clinical translation of these strategies are essential to advancing the next generation of lung cancer therapeutics.

Author Contributions

K.B.: writing original draft; M.A.: study design, conceptualization, writing, reviewing, and editing; M.Q.W.: reviewing. All authors have read and agreed to the published version of the manuscript.

Funding

No funding was received.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Li, C.; Lei, S.; Ding, L.; Xu, Y.; Wu, X.; Wang, H.; Zhang, Z.; Gao, T.; Zhang, Y.; Li, L. Global burden and trends of lung cancer incidence and mortality. Chin. Med. J. 2023, 136, 1583–1590. [Google Scholar] [CrossRef] [PubMed]
  3. Abbas, T.; Dutta, A. p21 in cancer: Intricate networks and multiple activities. Nat. Rev. Cancer 2009, 9, 400–414. [Google Scholar] [CrossRef] [PubMed]
  4. Zappa, C.; Mousa, S.A. Non-small cell lung cancer: Current treatment and future advances. Transl. Lung Cancer Res. 2016, 5, 288–300. [Google Scholar] [CrossRef]
  5. Chen, K.-C.; Tsai, S.-W.; Shie, R.-H.; Zeng, C.; Yang, H.-Y. Indoor air pollution increases the risk of lung cancer. Int. J. Environ. Res. Public Health 2022, 19, 1164. [Google Scholar] [CrossRef]
  6. Lancaster, H.L.; Heuvelmans, M.A.; Oudkerk, M. Low-dose computed tomography lung cancer screening: Clinical evidence and implementation research. J. Intern. Med. 2022, 292, 68–80. [Google Scholar] [CrossRef]
  7. Han, Z.; Aizezi, A.; Ma, L.; Su, Y.; Fan, L.; Liu, J. The association between human papillomavirus and lung cancer: A Mendelian randomization study. Infect. Genet. Evol. 2024, 123, 105646. [Google Scholar] [CrossRef]
  8. Osorio, J.C.; Candia-Escobar, F.; Corvalan, A.H.; Calaf, G.M.; Aguayo, F. High-risk human papillomavirus infection in lung cancer: Mechanisms and perspectives. Biology 2022, 11, 1691. [Google Scholar] [CrossRef]
  9. Xiong, W.-M.; Xu, Q.-P.; Li, X.; Xiao, R.-D.; Cai, L.; He, F. The association between human papillomavirus infection and lung cancer: A system review and meta-analysis. Oncotarget 2017, 8, 96419. [Google Scholar] [CrossRef]
  10. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  11. American Cancer Society. Global Cancer Facts & Figures, 5th ed.; American Cancer Society: Atlanta, GA, USA, 2024; Available online: https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/global-cancer-facts-and-figures/global-cancer-facts-and-figures-2024.pdf (accessed on 1 September 2025).
  12. Thun, M.J.; Carter, B.D.; Feskanich, D.; Freedman, N.D.; Prentice, R.; Lopez, A.D.; Hartge, P.; Gapstur, S.M. 50-year trends in smoking-related mortality in the United States. N. Engl. J. Med. 2013, 368, 351–364. [Google Scholar] [CrossRef]
  13. World Health Organization. WHO Report on the Global Tobacco Epidemic, 2025: Warning About the Dangers of Tobacco. 2025. Available online: https://www.who.int/publications/i/item/9789240112063 (accessed on 1 September 2025).
  14. Aredo, J.V.; Luo, S.J.; Gardner, R.M.; Sanyal, N.; Choi, E.; Hickey, T.P.; Riley, T.L.; Huang, W.-Y.; Kurian, A.W.; Leung, A.N. Tobacco smoking and risk of second primary lung cancer. J. Thorac. Oncol. 2021, 16, 968–979. [Google Scholar] [CrossRef]
  15. Samet, J.M.; Avila-Tang, E.; Boffetta, P.; Hannan, L.M.; Olivo-Marston, S.; Thun, M.J.; Rudin, C.M. Lung cancer in never smokers: Clinical epidemiology and environmental risk factors. Clin. Cancer Res. 2009, 15, 5626–5645. [Google Scholar] [CrossRef]
  16. Travis, R.C.; Allen, N.E.; Appleby, P.N.; Spencer, E.A.; Roddam, A.W.; Key, T.J. A prospective study of vegetarianism and isoflavone intake in relation to breast cancer risk in British women. Int. J. Cancer 2008, 122, 705–710. [Google Scholar] [CrossRef] [PubMed]
  17. Klaunig, J.E. Chemical carcinogenesis. In Casarett and Doull’s Toxicology: The Basic Science of Poisons, 8th ed.; McGraw-Hill Education: Columbus, OH, USA, 2014; p. 259. Available online: https://accesspharmacy.mhmedical.com/Content.aspx?bookid=958&sectionid=53483729 (accessed on 1 September 2025).
  18. O’Keeffe, L.M.; Taylor, G.; Huxley, R.R.; Mitchell, P.; Woodward, M.; Peters, S.A. Smoking as a risk factor for lung cancer in women and men: A systematic review and meta-analysis. BMJ Open 2018, 8, e021611. [Google Scholar] [CrossRef] [PubMed]
  19. Cohen, A.J.; Brauer, M.; Burnett, R.; Anderson, H.R.; Frostad, J.; Estep, K.; Balakrishnan, K.; Brunekreef, B.; Dandona, L.; Dandona, R. Estimates and 25-year trends of the global burden of disease attributable to ambient air pollution: An analysis of data from the Global Burden of Diseases Study 2015. Lancet 2017, 389, 1907–1918. [Google Scholar] [CrossRef] [PubMed]
  20. Liu, C.; Chen, R.; Sera, F.; Vicedo-Cabrera, A.M.; Guo, Y.; Tong, S.; Coelho, M.S.; Saldiva, P.H.; Lavigne, E.; Matus, P. Ambient particulate air pollution and daily mortality in 652 cities. N. Engl. J. Med. 2019, 381, 705–715. [Google Scholar] [CrossRef]
  21. Lin, S.-H.; Lee, H.-Y.; Chang, Y.-Y.; Jang, Y.; Chen, P.-C.; Wang, J.-D. Increased mortality risk for workers with a compensated, permanent occupational disability of the upper or lower extremities: A 21-year follow-up study. Am. J. Epidemiol. 2010, 171, 917–923. [Google Scholar] [CrossRef]
  22. Stayner, L.; Welch, L.S.; Lemen, R. The worldwide pandemic of asbestos-related diseases. Annu. Rev. Public Health 2013, 34, 205–216. [Google Scholar] [CrossRef]
  23. Rigamonti, A.; Viatore, M.; Polidori, R.; Rahal, D.; Erreni, M.; Fumagalli, M.R.; Zanini, D.; Doni, A.; Putignano, A.R.; Bossi, P. Integrating AI-Powered digital pathology and imaging mass cytometry identifies key classifiers of tumor cells, stroma, and immune cells in Non–Small cell lung Cancer. Cancer Res. 2024, 84, 1165–1177. [Google Scholar] [CrossRef]
  24. Sequeira, T.; Pinto, R.; Cardoso, C.; Almeida, C.; Aragão, R.; Almodovar, T.; Bicho, M.; Bicho, M.C.; Bárbara, C. HPV and Lung Cancer: A Systematic Review. Cancers 2024, 16, 3325. [Google Scholar] [CrossRef]
  25. Taniguchi, Y.; Matsuda, Y.; Mori, M.; Ito, M.; Ikari, T.; Tokoro, A.; Aiki, S.; Hoshino, S.; Kiuchi, D.; Suzuki, K. Effectiveness and safety of opioids for dyspnea in patients with lung cancer: Secondary analysis of multicenter prospective observational study. Transl. Lung Cancer Res. 2022, 11, 2395–2402. [Google Scholar] [CrossRef]
  26. Fois, S.S.; Paliogiannis, P.; Zinellu, A.; Fois, A.G.; Cossu, A.; Palmieri, G. Molecular epidemiology of the main druggable genetic alterations in non-small cell lung cancer. Int. J. Mol. Sci. 2021, 22, 612. [Google Scholar] [CrossRef]
  27. Reagan, M. Causes of cancer: Genetic, epigenetic, viral, microenvironmental, and environmental contributions to cancer. In Cancer: Prevention, Early Detection, Treatment and Recovery; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2019; pp. 53–74. [Google Scholar] [CrossRef]
  28. Travis, W.D.; Brambilla, E.; Nicholson, A.G.; Yatabe, Y.; Austin, J.H.; Beasley, M.B.; Chirieac, L.R.; Dacic, S.; Duhig, E.; Flieder, D.B. The 2015 World Health Organization classification of lung tumors: Impact of genetic, clinical and radiologic advances since the 2004 classification. J. Thorac. Oncol. 2015, 10, 1243–1260. [Google Scholar] [CrossRef] [PubMed]
  29. George, J.; Lim, J.S.; Jang, S.J.; Cun, Y.; Ozretić, L.; Kong, G.; Leenders, F.; Lu, X.; Fernández-Cuesta, L.; Bosco, G. Comprehensive genomic profiles of small cell lung cancer. Nature 2015, 524, 47–53. [Google Scholar] [CrossRef] [PubMed]
  30. Gay, C.M.; Stewart, C.A.; Park, E.M.; Diao, L.; Groves, S.M.; Heeke, S.; Nabet, B.Y.; Fujimoto, J.; Solis, L.M.; Lu, W. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities. Cancer Cell 2021, 39, 346–360.e7. [Google Scholar] [CrossRef]
  31. Pan, X.-B.; Chen, K.-H.; Huang, S.-T.; Jiang, Y.-M.; Ma, J.-L.; Liang, Z.-G.; Qu, S.; Li, L.; Chen, L.; Zhu, X.-D. Comparison of the efficacy between intensity-modulated radiotherapy and two-dimensional conventional radiotherapy in stage II nasopharyngeal carcinoma. Oncotarget 2017, 8, 78096–78104. [Google Scholar] [CrossRef]
  32. Skoulidis, F.; Goldberg, M.E.; Greenawalt, D.M.; Hellmann, M.D.; Awad, M.M.; Gainor, J.F.; Schrock, A.B.; Hartmaier, R.J.; Trabucco, S.E.; Gay, L. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 2018, 8, 822–835. [Google Scholar] [CrossRef]
  33. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef]
  34. Kamali, A.N.; Bautista, J.M.; Eisenhut, M.; Hamedifar, H. Immune checkpoints and cancer immunotherapies: Insights into newly potential receptors and ligands. Ther. Adv. Vaccines Immunother. 2023, 11, 25151355231192043. [Google Scholar] [CrossRef]
  35. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 pathways: Similarities, differences, and implications of their inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef] [PubMed]
  36. Shiravand, Y.; Khodadadi, F.; Kashani, S.M.A.; Hosseini-Fard, S.R.; Hosseini, S.; Sadeghirad, H.; Ladwa, R.; O’Byrne, K.; Kulasinghe, A. Immune checkpoint inhibitors in cancer therapy. Curr. Oncol. 2022, 29, 3044–3060. [Google Scholar] [CrossRef] [PubMed]
  37. Doroshow, D.B.; Sanmamed, M.F.; Hastings, K.; Politi, K.; Rimm, D.L.; Chen, L.; Melero, I.; Schalper, K.A.; Herbst, R.S. Immunotherapy in non–small cell lung cancer: Facts and hopes. Clin. Cancer Res. 2019, 25, 4592–4602. [Google Scholar] [CrossRef] [PubMed]
  38. Mamdani, H.; Matosevic, S.; Khalid, A.B.; Durm, G.; Jalal, S.I. Immunotherapy in lung cancer: Current landscape and future directions. Front. Immunol. 2022, 13, 823618. [Google Scholar] [CrossRef]
  39. Tan, S.; Li, D.; Zhu, X. Cancer immunotherapy: Pros, cons and beyond. Biomed. Pharmacother. 2020, 124, 109821. [Google Scholar] [CrossRef]
  40. Liu, C.; Yang, M.; Zhang, D.; Chen, M.; Zhu, D. Clinical cancer immunotherapy: Current progress and prospects. Front. Immunol. 2022, 13, 961805. [Google Scholar] [CrossRef]
  41. Acheampong, E.; Abed, A.; Morici, M.; Bowyer, S.; Amanuel, B.; Lin, W.; Millward, M.; S. Gray, E. Tumour PD-L1 expression in small-cell lung cancer: A systematic review and meta-analysis. Cells 2020, 9, 2393. [Google Scholar] [CrossRef]
  42. Caliman, E.; Fancelli, S.; Petroni, G.; Michelet, M.R.G.; Cosso, F.; Ottanelli, C.; Mazzoni, F.; Voltolini, L.; Pillozzi, S.; Antonuzzo, L. Challenges in the treatment of small cell lung cancer in the era of immunotherapy and molecular classification. Lung Cancer 2023, 175, 88–100. [Google Scholar] [CrossRef]
  43. National Cancer Institute. Testing the Addition of an Individualized Vaccine to Nab-Paclitaxel, Durvalumab and Tremelimumab and Chemotherapy in Patients with Metastatic Triple Negative Breast Cancer. ClinicalTrials.gov 2021. Available online: https://clinicaltrials.gov/study/NCT03606967 (accessed on 1 September 2025).
  44. The Lymphoma Academic Research Organisation; National Cancer Institute. Phase Ib Study of Intratumoral Selicrelumab (CD40 Agonist) in Combination with Atezolizumab in Refractory or Relapsed B-Cell Lymphoma. ClinicalTrials.gov 2019. Available online: https://clinicaltrials.gov/study/NCT03892525?cond=%22Hodgkin%27s%20Paragranuloma%22&intr=%22Atezolizumab%22&viewType=Table&rank=3 (accessed on 1 September 2025).
  45. Lin, A.; Huang, L.; Jiang, A.; Zhu, L.; Mou, W.; Li, Y.; Zhang, C.; Liu, Z.; Zhang, J.; Cheng, Q.; et al. Microbiota boost immuno-therapy? A meta-analysis dives into fecal microbiota transplantation and immune checkpoint inhibitors. BMC Med. 2025, 23, 341. [Google Scholar] [CrossRef]
  46. Synlogic. Safety and Tolerability of SYNB1891 Injection Alone or in Combination with Atezolizumab in Adult Subjects with Advanced/Metastatic Solid Tumors and Lymphoma. ClinicalTrials.gov 2021. Available online: https://www.mycancergenome.org/content/clinical_trials/NCT04167137/ (accessed on 1 September 2025).
  47. Grimmett, C.; Heneka, N.; Chambers, S. Psychological interventions prior to cancer surgery: A review of reviews. Curr. An-esthesiol. Rep. 2022, 12, 78–87. [Google Scholar] [CrossRef]
  48. Belli, C.; Trapani, D.; Viale, G.; D’Amico, P.; Duso, B.A.; Della Vigna, P.; Orsi, F.; Curigliano, G. Targeting the microenvi-ronment in solid tumors. Cancer Treat. Rev. 2018, 65, 22–32. [Google Scholar] [CrossRef] [PubMed]
  49. Vedenko, A.; Panara, K.; Goldstein, G.; Ramasamy, R.; Arora, H. Tumor microenvironment and nitric oxide: Concepts and mechanisms. In Tumor Microenvironment: Molecular Players–Part B; Springer: Cham, Switzerland, 2020; pp. 143–158. [Google Scholar] [CrossRef]
  50. Peng, M.; Mo, Y.; Wang, Y.; Wu, P.; Zhang, Y.; Xiong, F.; Guo, C.; Wu, X.; Li, Y.; Li, X. Neoantigen vaccine: An emerging tumor immunotherapy. Mol. Cancer 2019, 18, 128. [Google Scholar] [CrossRef] [PubMed]
  51. Jou, J.; Harrington, K.J.; Zocca, M.-B.; Ehrnrooth, E.; Cohen, E.E. The changing landscape of therapeutic cancer vaccines—Novel platforms and neoantigen identification. Clin. Cancer Res. 2021, 27, 689–703. [Google Scholar] [CrossRef] [PubMed]
  52. Muhammad, A.; Champeimont, J.; Mayr, U.B.; Lubitz, W.; Kudela, P. Bacterial ghosts as carriers of protein subunit and DNA-encoded antigens for vaccine applications. Expert Rev. Vaccines 2012, 11, 97–116. [Google Scholar] [CrossRef]
  53. Anwer, M.; Bhaliya, K.; Munn, A.; Wei, M.Q. Bacterial ghosts: A breakthrough approach to cancer vaccination. Biomed. Pharmacother. 2025, 182, 117766. [Google Scholar] [CrossRef]
  54. Lawler, S.E.; Speranza, M.-C.; Cho, C.-F.; Chiocca, E.A. Oncolytic Viruses in Cancer Treatment: A Review. JAMA Oncol. 2017, 3, 841–849. [Google Scholar] [CrossRef]
  55. My Cancer Genome. Trial with BNT111 and Cemiplimab in Combination or as Single Agents in Patients with An-ti-PD1-Refractory/Relapsed, Unresectable Stage III or IV Melanoma. ClinicalTrials.gov 2021. Available online: https://www.mycancergenome.org/content/clinical_trials/NCT04526899/ (accessed on 1 September 2025).
  56. AbbVie. A Phase 1, Multicenter, Open-Label Study to Determine the Safety, Tolerability, Pharmacokinetics, and Preliminary Efficacy of ABBV-927 with ABBV-368, Budigalimab (ABBV-181), and/or Chemotherapy in Subjects with Locally Advanced or Metastatic Solid Tumors. ClinicalTrials.gov 2019. Available online: https://clinicaltrials.gov/study/NCT03893955 (accessed on 1 September 2025).
  57. Buonaguro, L.; Tornesello, M.L.; Buonaguro, F.M. Cancer vaccine development: From preclinical models to clinical trials. Vaccines 2020, 8, 402. [Google Scholar] [CrossRef]
  58. Roswell Park Cancer Institute; National Cancer Institute. FGFR1 Amplification as A Predictor of Efficacy in a Biomarker-Driven Phase II Study of BIBF 1120 in Advanced Squamous Cell Lung Cancer Patients Who Have Failed Up to Two Prior Chemotherapeutic Regimens. ClinicalTrials.gov 2013. Available online: https://www.clinicaltrials.gov/study/NCT01948141 (accessed on 1 September 2025).
  59. Nemunaitis, J.; Dillman, R.O.; Schwarzenberger, P.O.; Senzer, N.; Cunningham, C.; Cutler, J.; Tong, A.; Kumar, P.; Pappen, B.; Hamilton, C. Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene-modified allogeneic tumor cell vaccine in non–small-cell lung cancer. J. Clin. Oncol. 2006, 24, 4721–4730. [Google Scholar] [CrossRef]
  60. Valanparambil, R.M.; Carlisle, J.; Linderman, S.L.; Akthar, A.; Millett, R.L.; Lai, L.; Chang, A.; McCook, A.A.; Switchenko, J.; Nasti, T.H. Antibody response to SARS-CoV-2 mRNA vaccine in lung cancer patients: Reactivity to vaccine antigen and variants of concern. MedRxiv 2022, 40, 3808–3816. [Google Scholar] [CrossRef]
  61. Kreiter, S.; Selmi, A.; Diken, M.; Koslowski, M.; Britten, C.M.; Huber, C.; Türeci, Ö.; Sahin, U. Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity. Cancer Res. 2010, 70, 9031–9040. [Google Scholar] [CrossRef]
  62. Rezaei, T.; Davoudian, E.; Khalili, S.; Amini, M.; Hejazi, M.; de la Guardia, M.; Mokhtarzadeh, A. Strategies in DNA vaccine for melanoma cancer. Pigment. Cell Melanoma Res. 2021, 34, 869–891. [Google Scholar] [CrossRef]
  63. Hobernik, D.; Bros, M. DNA vaccines—How far from clinical use? Int. J. Mol. Sci. 2018, 19, 3605. [Google Scholar] [CrossRef] [PubMed]
  64. Gan, Y.; Li, X.; Han, S.; Liang, Q.; Ma, X.; Rong, P.; Wang, W.; Li, W. The cGAS/STING pathway: A novel target for cancer therapy. Front. Immunol. 2022, 12, 795401. [Google Scholar] [CrossRef] [PubMed]
  65. Ishikawa, H.; Ma, Z.; Barber, G.N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 2009, 461, 788–792. [Google Scholar] [CrossRef] [PubMed]
  66. Lopes, A.; Vanvarenberg, K.; Préat, V.; Vandermeulen, G. Codon-optimized P1A-encoding DNA vaccine: Toward a therapeutic vaccination against P815 mastocytoma. Mol. Ther. Nucleic Acids 2017, 8, 404–415. [Google Scholar] [CrossRef]
  67. Gupta, M.; Wahi, A.; Sharma, P.; Nagpal, R.; Raina, N.; Kaurav, M.; Bhattacharya, J.; Rodrigues Oliveira, S.M.; Dolma, K.G.; Paul, A.K. Recent advances in cancer vaccines: Challenges, achievements, and futuristic prospects. Vaccines 2022, 10, 2011. [Google Scholar] [CrossRef]
  68. Abd-Aziz, N.; Poh, C.L. Development of peptide-based vaccines for cancer. J. Oncol. 2022, 2022, 9749363. [Google Scholar] [CrossRef]
  69. Li, W.; Joshi, M.D.; Singhania, S.; Ramsey, K.H.; Murthy, A.K. Peptide vaccine: Progress and challenges. Vaccines 2014, 2, 515–536. [Google Scholar] [CrossRef]
  70. Anwer, M.; Wei, M.Q. Harnessing the power of probiotic strains in functional foods: Nutritive, therapeutic, and next-generation challenges. Food Sci.Biotechnol. 2024, 33, 2081–2095. [Google Scholar] [CrossRef]
  71. Paston, S.J.; Brentville, V.A.; Symonds, P.; Durrant, L.G. Cancer vaccines, adjuvants, and delivery systems. Front. Immunol. 2021, 12, 627932. [Google Scholar] [CrossRef]
  72. Cuzzubbo, S.; Mangsbo, S.; Nagarajan, D.; Habra, K.; Pockley, A.G.; McArdle, S.E. Cancer vaccines: Adjuvant potency, importance of age, lifestyle, and treatments. Front. Immunol. 2021, 11, 615240. [Google Scholar] [CrossRef]
  73. Luo, M.; Wang, H.; Wang, Z.; Cai, H.; Lu, Z.; Li, Y.; Du, M.; Huang, G.; Wang, C.; Chen, X. A STING-activating nanovaccine for cancer immunotherapy. Nat. Nanotechnol. 2017, 12, 648–654. [Google Scholar] [CrossRef]
  74. Gulen, M.F.; Koch, U.; Haag, S.M.; Schuler, F.; Apetoh, L.; Villunger, A.; Radtke, F.; Ablasser, A. Signalling strength determines proapoptotic functions of STING. Nat. Commun. 2017, 8, 427. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, J.-J.; Li, W.-H.; Chen, P.-G.; Zhang, B.-D.; Hu, H.-G.; Li, Q.-Q.; Zhao, L.; Chen, Y.-X.; Zhao, Y.-F.; Li, Y.-M. Targeting STING with cyclic di-GMP greatly augmented immune responses of glycopeptide cancer vaccines. Chem. Commun. 2018, 54, 9655–9658. [Google Scholar] [CrossRef]
  76. Shao, Y.; Sun, Z.-Y.; Wang, Y.; Zhang, B.-D.; Liu, D.; Li, Y.-M. Designable immune therapeutical vaccine system based on DNA supramolecular hydrogels. ACS Appl. Mater. Interfaces 2018, 10, 9310–9314. [Google Scholar] [CrossRef] [PubMed]
  77. Vonderheide, R.H.; Glennie, M.J. Agonistic CD40 antibodies and cancer therapy. Clin. Cancer Res. 2013, 19, 1035–1043. [Google Scholar] [CrossRef]
  78. Nimanong, S.; Ostroumov, D.; Wingerath, J.; Knocke, S.; Woller, N.; Gürlevik, E.; Falk, C.S.; Manns, M.P.; Kühnel, F.; Wirth, T.C. CD40 signaling drives potent cellular immune responses in heterologous cancer vaccinations. Cancer Res. 2017, 77, 1918–1926. [Google Scholar] [CrossRef]
  79. Chandra, D.; Quispe-Tintaya, W.; Jahangir, A.; Asafu-Adjei, D.; Ramos, I.; Sintim, H.O.; Zhou, J.; Hayakawa, Y.; Karaolis, D.K.; Gravekamp, C. STING ligand c-di-GMP improves cancer vaccination against metastatic breast cancer. Cancer Immunol. Res. 2014, 2, 901–910. [Google Scholar] [CrossRef]
  80. Sun, B.; Xia, T. Nanomaterial-based vaccine adjuvants. J. Mater. Chem. B 2016, 4, 5496–5509. [Google Scholar] [CrossRef]
  81. Wang, M.; Sharma, A.; Osazuwa-Peters, N.; Simpson, M.C.; Schootman, M.; Piccirillo, J.F.; Huh, W.K.; Boakye, E.A. Risk of subsequent malignant neoplasms after an index potentially-human papillomavirus (HPV)-associated cancers. Cancer Epidemiol. 2020, 64, 101649. [Google Scholar] [CrossRef]
  82. Zhang, R.; Wang, C.; Guan, Y.; Wei, X.; Sha, M.; Yi, M.; Jing, M.; Lv, M.; Guo, W.; Xu, J. Manganese salts function as potent adjuvants. Cell Mol. Immunol. 2021, 18, 1222–1234. [Google Scholar] [CrossRef]
  83. Xia, Y.; Xie, Y.; Yu, Z.; Xiao, H.; Jiang, G.; Zhou, X.; Yang, Y.; Li, X.; Zhao, M.; Li, L. The mevalonate pathway is a druggable target for vaccine adjuvant discovery. Cell 2018, 175, 1059–1073.e21. [Google Scholar] [CrossRef]
  84. Ma, X.; Wang, Y.; Zhao, T.; Li, Y.; Su, L.-C.; Wang, Z.; Huang, G.; Sumer, B.D.; Gao, J. Ultra-pH-sensitive nanoprobe library with broad pH tunability and fluorescence emissions. J. Am. Chem. Soc. 2014, 136, 11085–11092. [Google Scholar] [CrossRef]
  85. Horning, K.J.; Caito, S.W.; Tipps, K.G.; Bowman, A.B.; Aschner, M. Manganese is essential for neuronal health. Annu. Rev. Nutr. 2015, 35, 71–108. [Google Scholar] [CrossRef]
  86. Bando, J.K.; Colonna, M. Group 2 innate lymphoid cells induce antibody production in gastric tissue. Trends Immunol. 2020, 41, 643–645. [Google Scholar] [CrossRef]
  87. Duan, T.; Du, Y.; Xing, C.; Wang, H.Y.; Wang, R.-F. Toll-like receptor signaling and its role in cell-mediated immunity. Front. Immunol. 2022, 13, 812774. [Google Scholar] [CrossRef]
  88. Asami, J.; Shimizu, T. Structural and functional understanding of the toll-like receptors. Protein Sci. 2021, 30, 761–772. [Google Scholar] [CrossRef]
  89. Finn, O.J. The dawn of vaccines for cancer prevention. Nat. Rev. Immunol. 2018, 18, 183–194. [Google Scholar] [CrossRef] [PubMed]
  90. Cho, J.H.; Lee, H.-J.; Ko, H.-J.; Yoon, B.-I.; Choe, J.; Kim, K.-C.; Hahn, T.-W.; Han, J.A.; Choi, S.S.; Jung, Y.M. The TLR7 agonist imiquimod induces anti-cancer effects via autophagic cell death and enhances anti-tumoral and systemic immunity during radiotherapy for melanoma. Oncotarget 2017, 8, 24932. [Google Scholar] [CrossRef] [PubMed]
  91. Dajon, M.; Iribarren, K.; Cremer, I. Toll-like receptor stimulation in cancer: A pro-and anti-tumor double-edged sword. Immunobiology 2017, 222, 89–100. [Google Scholar] [CrossRef] [PubMed]
  92. Dyavar, S.R.; Singh, R.; Emani, R.; Pawar, G.P.; Chaudhari, V.D.; Podany, A.T.; Avedissian, S.N.; Fletcher, C.V.; Salunke, D.B. Role of toll-like receptor 7/8 pathways in regulation of interferon response and inflammatory mediators during SARS-CoV2 infection and potential therapeutic options. Biomed. Pharmacother. 2021, 141, 111794. [Google Scholar] [CrossRef]
  93. Sartorius, R.; Trovato, M.; Manco, R.; D’Apice, L.; De Berardinis, P. Exploiting viral sensing mediated by Toll-like receptors to design innovative vaccines. npj Vaccines 2021, 6, 127. [Google Scholar] [CrossRef] [PubMed]
  94. Lacarrubba, F.; Nasca, M.R.; Micali, G. Advances in the use of topical imiquimod to treat dermatologic disorders. Ther. Clin. Risk Manag. 2008, 4, 87–97. [Google Scholar] [CrossRef] [PubMed]
  95. Bhaliya, K.R.; Anwer, M.; Munn, A.; Wei, M.Q. New horizons in cancer immunotherapy: The evolving role of R848 and R837. Mol. Clin. Oncol. 2024, 22, 4. [Google Scholar] [CrossRef] [PubMed]
  96. Zhou, Y.; Chen, X.; Cao, Z.; Li, J.; Long, H.; Wu, Y.; Zhang, Z.; Sun, Y. R848 is involved in the antibacterial immune response of golden pompano (Trachinotus ovatus) through TLR7/8-MyD88-NF-κb-signaling pathway. Front. Immunol. 2021, 11, 617522. [Google Scholar] [CrossRef]
  97. Bhagchandani, S.; Johnson, J.A.; Irvine, D.J. Evolution of Toll-like receptor 7/8 agonist therapeutics and their delivery approaches: From antiviral formulations to vaccine adjuvants. Adv. Drug Deliv. Rev. 2021, 175, 113803. [Google Scholar] [CrossRef]
  98. Alam, M.M.; Yang, D.; Trivett, A.; Meyer, T.J.; Oppenheim, J.J. HMGN1 and R848 synergistically activate dendritic cells using multiple signaling pathways. Front. Immunol. 2018, 9, 2982. [Google Scholar] [CrossRef]
  99. Ott, P.A.; Hu-Lieskovan, S.; Chmielowski, B.; Govindan, R.; Naing, A.; Bhardwaj, N.; Margolin, K.; Awad, M.M.; Hellmann, M.D.; Lin, J.J. A phase Ib trial of personalized neoantigen therapy plus anti-PD-1 in patients with advanced melanoma, non-small cell lung cancer, or bladder cancer. Cell 2020, 183, 347–362.e24. [Google Scholar] [CrossRef]
  100. Wu, D.-W.; Jia, S.-P.; Xing, S.-J.; Ma, H.-l.; Wang, X.; Tang, Q.-Y.; Li, Z.-W.; Wu, Q.; Bai, M.; Zhang, X.-Y. Personalized neoantigen cancer vaccines: Current progression, challenges and a bright future. Clin. Exp. Med. 2024, 24, 229. [Google Scholar] [CrossRef]
  101. Weber, J.S.; Carlino, M.S.; Khattak, A.; Meniawy, T.; Ansstas, G.; Taylor, M.H.; Kim, K.B.; McKean, M.; Long, G.V.; Sullivan, R.J. Individualised neoantigen therapy mRNA-4157 (V940) plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma (KEYNOTE-942): A randomised, phase 2b study. Lancet 2024, 403, 632–644. [Google Scholar] [CrossRef]
  102. Braun, D.A.; Moranzoni, G.; Chea, V.; McGregor, B.A.; Blass, E.; Tu, C.R.; Vanasse, A.P.; Forman, C.; Forman, J.; Afeyan, A.B. A neoantigen vaccine generates antitumour immunity in renal cell carcinoma. Nature 2025, 639, 474–482. [Google Scholar] [CrossRef] [PubMed]
  103. Al Fayez, N.; Nassar, M.S.; Alshehri, A.A.; Alnefaie, M.K.; Almughem, F.A.; Alshehri, B.Y.; Alawad, A.O.; Tawfik, E.A. Recent advancement in mRNA vaccine development and applications. Pharmaceutics 2023, 15, 1972. [Google Scholar] [CrossRef] [PubMed]
  104. Bottomly, D.; Long, N.; Schultz, A.R.; Kurtz, S.E.; Tognon, C.E.; Johnson, K.; Abel, M.; Agarwal, A.; Avaylon, S.; Benton, E. Integrative analysis of drug response and clinical outcome in acute myeloid leukemia. Cancer Cell 2022, 40, 850–864.e9. [Google Scholar] [CrossRef] [PubMed]
  105. Chiocca, E.A.; Rabkin, S.D. Oncolytic viruses and their application to cancer immunotherapy. Cancer Immunol. Res. 2014, 2, 295–300. [Google Scholar] [CrossRef]
  106. Wang, Z.; Huang, D.; Li, S.; Ke, L. Role of tumor mutational burden in patients with urothelial carcinoma treated with immune checkpoint inhibitors: A systematic review and meta-analysis. Front. Immunol. 2025, 16, 1592761. [Google Scholar] [CrossRef]
  107. Foy, S.P.; Jacoby, K.; Bota, D.A.; Hunter, T.; Pan, Z.; Stawiski, E.; Ma, Y.; Lu, W.; Peng, S.; Wang, C.L. Non-viral precision T cell receptor replacement for personalized cell therapy. Nature 2023, 615, 687–696. [Google Scholar] [CrossRef]
  108. Klempner, S.J.; Fabrizio, D.; Bane, S.; Reinhart, M.; Peoples, T.; Ali, S.M.; Sokol, E.S.; Frampton, G.; Schrock, A.B.; Anhorn, R. Tumor mutational burden as a predictive biomarker for response to immune checkpoint inhibitors: A review of current evidence. Oncologist 2020, 25, e147–e159. [Google Scholar] [CrossRef]
Figure 1. Global distribution of new cancer cases by type according to GLOBOCAN 2020. Lung cancer remains the primary burden in new cancer cases among all other cancers.
Figure 1. Global distribution of new cancer cases by type according to GLOBOCAN 2020. Lung cancer remains the primary burden in new cancer cases among all other cancers.
Vaccines 13 00957 g001
Figure 2. Mechanism of HPV-mediated lung cancer. The figure outlines how high-risk HPV infects lung epithelial cells and expresses E6 and E7 oncoproteins, which disrupt p53 and Rb tumor suppressor pathways. This leads to genomic instability, uncontrolled proliferation, and progression through various stages of tumor development, ultimately resulting in metastasis.
Figure 2. Mechanism of HPV-mediated lung cancer. The figure outlines how high-risk HPV infects lung epithelial cells and expresses E6 and E7 oncoproteins, which disrupt p53 and Rb tumor suppressor pathways. This leads to genomic instability, uncontrolled proliferation, and progression through various stages of tumor development, ultimately resulting in metastasis.
Vaccines 13 00957 g002
Figure 3. Comprehensive overview of treatment modalities for cancer.
Figure 3. Comprehensive overview of treatment modalities for cancer.
Vaccines 13 00957 g003
Figure 4. Mechanisms of immunotherapy in lung cancer. Checkpoint inhibitors, such as anti-PD-1, anti-PD-L1, and anti-CTLA-4, reactivate exhausted T cells by blocking inhibitory immune checkpoints. This enhances cytotoxic T cell function and promotes tumor cell elimination within the immunosuppressive tumor microenvironment.
Figure 4. Mechanisms of immunotherapy in lung cancer. Checkpoint inhibitors, such as anti-PD-1, anti-PD-L1, and anti-CTLA-4, reactivate exhausted T cells by blocking inhibitory immune checkpoints. This enhances cytotoxic T cell function and promotes tumor cell elimination within the immunosuppressive tumor microenvironment.
Vaccines 13 00957 g004
Table 1. Challenges in immunotherapy and solutions along with ongoing clinical trials.
Table 1. Challenges in immunotherapy and solutions along with ongoing clinical trials.
ChallengeDescriptionProposed Solutions/StrategiesOngoing TrialsReferences
Lack of predictive biomarkersPD-L1 and TMB are inconsistent predictors of ICI responseComposite biomarkers: gene expression, peripheral immune profilingNCT03606967: multiomic biomarker analysis in NSCLC[43]
Primary resistance“Cold” tumors lack immune infiltrationCombination with chemo, radiotherapy, or STING agonists to enhance immunogenicityNCT03892525: STING agonist + PD-1 blockade in solid tumors[44]
Acquired resistanceTumor escapes after initial ICI responseDual checkpoint blockade (e.g., PD-1 + LAG-3), epigenetic therapyNCT03686202: anti–PD-1 + LAG-3 in resistant NSCLC[45]
Immune-related adverse eventsAutoimmunity affecting skin, GI, lungs, etc.IL-6 blockades, corticosteroids, better screening algorithmsNCT04167137: tocilizumab for irAEs[46]
Table 2. Cancer vaccines platforms in early developmental stage.
Table 2. Cancer vaccines platforms in early developmental stage.
Development StageVaccine TypeMechanism of ActionExamples/StatusReferences
PreclinicalBacterial ghost vaccinesEmpty bacterial envelopes deliver tumor antigens, enhance innate immunityAnimal models show promise in lung cancer[52,53]
Oncolytic virus-based vaccinesSelectively replicate in tumors, trigger immune responseAdenovirus and HSV-based vaccines under development[54]
Early-phase clinical trialsmRNA vaccinesEncode neoantigens, induce antigen-specific T cell responseNCT04526899: personalized mRNA vaccines in NSCLC[55]
DNA vaccinesPlasmid DNA delivered by electroporation induces immune responseNCT03893955: DNA vaccine targeting surviving[56]
Peptide-based vaccinesTumor-associated antigens presented on MHC moleculesNCT04397900: multi-epitope vaccine trials[57]
Advanced/approvedDendritic cell (DC) vaccinesAutologous DCs loaded with tumor antigens and re-infusedNCT01948141: DC vaccine with chemo in NSCLC[58]
Allogeneic whole-cell vaccinesGenetically modified tumor cells as broad antigen sourceBelagenpumatucel-L in Phase III trials (not FDA-approved)[59]
Table 3. Mechanism of action and clinical status of traditional and emerging adjuvants.
Table 3. Mechanism of action and clinical status of traditional and emerging adjuvants.
CategoryAdjuvant/AgentMechanism of ActionClinical Status/NotesReferences
Traditional adjuvantsTLR agonists (CpG ODN, imiquimod, MPL)Activate pattern recognition receptors (TLR7, TLR9, TLR4), enhance dendritic cell maturation, type I IFN secretionWidely used in cancer vaccines; imiquimod FDA-approved for topical use; effective in boosting immune response[71,72]
Aluminum saltsPromote inflammasome activation, support humoral immunityCommon in preventive vaccines; less effective in eliciting cytotoxic T cell responses[71]
Poly-ICLCSynthetic dsRNA analog activates TLR3 and MDA5 pathways, stimulates cytokine productionUsed in early-stage cancer vaccine trials; boosts T cell activation[71]
Emerging adjuvantsSTING agonists (cGAMP, CDNs)Stimulate cytosolic DNA sensing pathways, induce strong type I IFN responseEarly-phase clinical trials; potent activators of antitumor immunity but require safety optimization[73,74]
CD40 agonistsActivate dendritic cells and B cells, enhance antigen cross-presentationPromising in combination therapies; clinical use limited by toxicity concerns[67]
Cytokines (GM-CSF)Recruit and activate dendritic cells at vaccination sitesMixed clinical results; potential to expand immunosuppressive cells under some conditions[76,77]
Inorganic nanoparticles (TiO2, Mn-based)Enhance antigen cross-presentation, modulate STING pathwayPreclinical stage; promising delivery platforms with immunomodulatory effects[80,82]
Imidazoquinolines (IMDs)Imiquimod (R837), resiquimod (R848)TLR7/8 agonists; induce NF-κB and IRF activation, promote IL-12 and type I IFN productionImiquimod FDA-approved for topical cancers; resiquimod in clinical trials; limited by systemic toxicity[95,96]
Table 4. Clinical feasibility and timeline of immunotherapeutic strategies.
Table 4. Clinical feasibility and timeline of immunotherapeutic strategies.
Immunotherapy StrategyCurrent StageEstimated Clinical TimelineNotes
Personalized neoantigen vaccinesEarly clinical trials3–7 yearsmRNA and peptide vaccines showing promising immune responses and manageable safety profiles.
Combination with immune checkpoint inhibitorsClinical trials/ongoing use3–5 yearsSynergistic effects are well documented, rapidly moving toward broader clinical adoption.
Viral vector and oncolytic virus platformsPreclinical/early trials5–10 yearsHigh potential but requires optimization for safety and efficacy.
STING and CD40 agonists as adjuvantsPreclinical/Phase I trials5–10+ yearsPromising immune activation, but toxicity and delivery need refinement.
Bacterial ghost and nanoparticle-based vaccinesPreclinical7–15 yearsInnovative delivery systems with encouraging results in animal models; translation pending.
Predictive biomarker developmentEarly clinical/exploratory3–7 yearsBiomarkers for patient stratification improving vaccine personalization and response tracking.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bhaliya, K.; Anwer, M.; Wei, M.Q. Emerging Therapeutic Strategies for Lung Cancer: The Role of Immunotherapy and HPV-Targeted Cancer Vaccines. Vaccines 2025, 13, 957. https://doi.org/10.3390/vaccines13090957

AMA Style

Bhaliya K, Anwer M, Wei MQ. Emerging Therapeutic Strategies for Lung Cancer: The Role of Immunotherapy and HPV-Targeted Cancer Vaccines. Vaccines. 2025; 13(9):957. https://doi.org/10.3390/vaccines13090957

Chicago/Turabian Style

Bhaliya, Krupa, Muneera Anwer, and Ming Q. Wei. 2025. "Emerging Therapeutic Strategies for Lung Cancer: The Role of Immunotherapy and HPV-Targeted Cancer Vaccines" Vaccines 13, no. 9: 957. https://doi.org/10.3390/vaccines13090957

APA Style

Bhaliya, K., Anwer, M., & Wei, M. Q. (2025). Emerging Therapeutic Strategies for Lung Cancer: The Role of Immunotherapy and HPV-Targeted Cancer Vaccines. Vaccines, 13(9), 957. https://doi.org/10.3390/vaccines13090957

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop