Previous Article in Journal
Dermatophilosis: Current Advances and Future Directions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genome Editing Against HPV-Driven Cancers: From Bench to Clinic

1
School of Life Sciences, Pharmacy and Chemistry, Kingston University London, London KT1 2EE, UK
2
School of Medicine, Koc University, Sarıyer, Istanbul 34450, Turkey
3
Department of Biochemistry, School of Medicine & Genetic and Metabolic Disease Research and Investigation Center, Marmara University, Istanbul 34854, Turkey
4
Department of Biochemistry, School of Medicine, Recep Tayyip Erdogan University, Rize 53100, Turkey
5
Division of Medical Oncology, Memorial Hospital, Antalya 07050, Turkey
*
Author to whom correspondence should be addressed.
Acta Microbiol. Hell. 2025, 70(4), 41; https://doi.org/10.3390/amh70040041
Submission received: 2 August 2025 / Revised: 9 September 2025 / Accepted: 9 October 2025 / Published: 31 October 2025

Abstract

Genome editing technologies, including CRISPR/Cas9, TALENs, and ZFNs, offer promising approaches to disrupt HPV oncogenes E6 and E7, thereby restoring tumor-suppressor pathways. In this review, we summarize recent preclinical findings demonstrating selective apoptosis and tumor regression in HPV-positive cell and animal models, as well as early-phase clinical studies exploring local CRISPR-based therapies. We also compare the relative strengths and limitations of major editing platforms, discuss delivery strategies, and highlight their potential integration with immunotherapy and conventional treatments. While preclinical studies show encouraging efficacy (e.g., up to 60% tumor regression in xenograft models and marked reactivation of p53/pRb pathways), translation into routine practice remains limited by challenges such as efficient delivery, minimizing off-target effects, long-term safety, cost, and ethical considerations. Continued optimization of high-fidelity nucleases, tissue-specific delivery systems, and genotype-tailored guide RNAs will be essential. Genome editing therefore represents a potential future addition to the therapeutic landscape of HPV-related diseases, but substantial barriers must be addressed before clinical implementation.

1. Introduction

Human papillomavirus (HPV) is the primary etiological factor in cervical cancer and a significant contributor to anogenital and oropharyngeal malignancies worldwide [1,2,3]. Although prophylactic vaccines have proven highly effective in preventing new infections, they provide no therapeutic benefit to individuals already infected or with established lesions [4,5]. Similarly, cytology-based screening and surgical excision have reduced cancer incidence in high-resource settings but remain less accessible in low- and middle-income countries, where the burden is highest [6]. For patients with persistent HPV infection or precancerous lesions, current management strategies are limited, often oscillating between overtreatment and long-term surveillance, neither of which eliminates the underlying viral reservoir [7].
These therapeutic gaps underscore the urgent need for novel strategies that directly target HPV’s oncogenic drivers. Genome editing technologies—including CRISPR/Cas9, transcription activator-like effector nucleases (TALENs), and zinc-finger nucleases (ZFNs)—offer such an approach by disrupting viral oncogenes (E6/E7) or excising integrated viral DNA [8,9,10]. Unlike conventional modalities, these tools have the potential to eliminate the molecular basis of HPV-driven carcinogenesis. Recent preclinical studies in cell and animal models provide encouraging proof-of-concept, while early-phase clinical efforts are beginning to assess safety and feasibility in humans [11,12,13].
This review aims to critically evaluate the current state of HPV-targeted genome editing. We focus on mechanisms, preclinical and clinical progress, integration into existing oncological treatments, and the formidable challenges that must be addressed—including delivery barriers, off-target risks, ethical considerations, and cost. By outlining both opportunities and limitations, we seek to provide a balanced overview of this rapidly evolving field.

2. Methods

This review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines to ensure transparency and reproducibility.

2.1. Literature Search Strategy

A systematic literature search was performed in PubMed, Web of Science, and Scopus databases for studies published between January 2010 and June 2024. The following keywords and Boolean operators were used:
(“HPV” OR “Human papillomavirus”) AND (“CRISPR” OR “TALEN” OR “ZFN” OR “genome editing” OR “gene editing”) AND (“therapy” OR “treatment” OR “clinical trial” OR “preclinical”).
Reference lists of relevant reviews and primary studies were also manually screened to identify additional eligible publications.

2.2. Inclusion and Exclusion Criteria

Inclusion criteria:
  • Original in vitro, in vivo, or clinical studies investigating genome editing technologies (CRISPR, TALENs, ZFNs) in HPV infection or HPV-related cancers.
  • Studies reporting mechanistic, therapeutic, or translational outcomes.
  • Articles published in English.
Exclusion criteria:
  • Non-HPV related studies.
  • Editorials, commentaries, and conference abstracts without primary data.
  • Duplicate publications.

2.3. Study Selection and Data Extraction

Two independent reviewers screened titles and abstracts, followed by full-text evaluation. Disagreements were resolved by consensus. Data extracted included study design, model system (cell line, animal model, clinical cohort), editing platform, target genes, delivery system, and outcomes.

2.4. PRISMA Flow Diagram

A PRISMA flow diagram (Figure 1) summarizes the study selection process. The database search (January 2010–June 2024) identified 1248 records from databases and 42 additional records from manual searches. After removal of 290 duplicates, 1000 records were screened by title and abstract, of which 820 were excluded. Full-text assessment was conducted for 180 articles, with 120 excluded for not meeting the inclusion criteria. Finally, 60 studies were included in this review. In total, 60 eligible primary studies (in vitro, in vivo, and clinical) were included in this systematic review. Additional background and methodological references were cited where relevant, resulting in 130 references overall in the manuscript.

3. Genome Editing Technologies

Modern genome editing relies on programmable nucleases that create targeted DNA double-strand breaks (DSBs), triggering cellular DNA repair processes that can introduce mutations or integrate new DNA [3,14,15].The three primary platforms are zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-associated nuclease). All three systems ultimately achieve gene editing via the same mechanism—DSB induction and the cell’s subsequent repair by non-homologous end joining (NHEJ) or homology-directed repair (HDR) but they differ in how they recognize DNA targets and, in their practicality, and precision.

3.1. CRISPR/Cas9

The CRISPR/Cas9 system, adapted from a bacterial adaptive immune mechanism, has revolutionized genome editing with its simplicity and efficiency [2]. In CRISPR/Cas9, a programmable single-guide RNA directs the Cas9 endonuclease to a complementary DNA sequence, where Cas9 introduces a DSB. Designing a CRISPR experiment is as straightforward as changing the 20-nucleotide guide sequence in the sgRNA, in contrast to the protein engineering required for ZFNs and TALENs. This RNA-guided flexibility makes CRISPR extremely versatile and fast for testing many targets *****. CRISPR can also deliver multiple sgRNAs to target several sites simultaneously, which is useful for knocking out both E6 and E7 oncogenes together or multiple viral genome sites in one go [4].
CRISPR’s main advantages are its ease of design, high efficiency, and scalability. Thousands of guides can be cheaply synthesized and tested, enabling large genetic screens or personalized target adaptation. In HPV research, the ability to quickly craft sgRNAs for different HPV genotypes (e.g., HPV16 vs. HPV18) means therapies can be tailored to the patient’s specific virus [5]. Several studies have reported that wild-type Cas9 can induce indels in the majority of target HPV DNA sequences in a cell population [16]. Moreover, CRISPR has been leveraged to reactivate silenced tumor suppressor pathways in HPV-positive cancer cells—for example, by knocking out E6, thereby freeing p53, or knocking out E7 to restore pRB function [8]. Another advantage in the HPV context is that CRISPR can target integrated viral DNA within the host genome, conventional antivirals or RNA interference cannot permanently do [17].
The major concern for CRISPR/Cas9 is off-target activity, where the Cas9-sgRNA complex cuts at unintended genomic sites that resemble the target sequence. Off-target mutations could potentially inactivate other genes or cause genomic instability—a significant safety issue for any therapeutic application. However, off-target profiles can be improved by using high-fidelity Cas9 variants and by careful guide sequence design using bioinformatic tools. Indeed, bioinformatic improvements have largely mitigated off-target risks by enabling the selection of highly specific sgRNAs and by providing methods to empirically detect off-target cuts (e.g., GUIDE-seq) [10,18]. Another limitation is delivery. the SpCas9 protein (≈160 kDa) plus its sgRNA (≈100 nt) are large in size, making it challenging to package into some vectors (for instance, the popular AAV vectors have a ~4.7 kb cargo limit, which just accommodates Cas9 and a short sgRNA). Various studies explored smaller Cas9 orthologs (like SaCas9) or transient mRNA/protein delivery to address this [19,20]. There is also the possibility of immune responses against Cas9, since it is a foreign bacterial protein; antibodies to Cas9 have been detected in humans, presumably from prior exposure to Cas9-bearing bacteria, but strategies like transient local delivery or using human-compatible delivery vehicles aim to minimize this risk [21]. Overall, CRISPR/Cas9 has proven to be a game-changing tool, combining relative precision with unparalleled programmability, which is why it has become the preferred genome editing method in recent HPV research and therapeutic development.

3.2. TALENs

TALENs were one of the first “user-friendly” genome editing systems, developed shortly before CRISPR. A TALEN is composed of a DNA-binding domain built from TALE repeats (derived from plant pathogen transcription activator-like effectors) fused to the FokI nuclease domain. Each TALE repeat is a 33–35 amino acid module that recognizes one base pair via two hypervariable residues (repeat-variable diresidues, RVDs). By assembling ~15–20 repeats with appropriate RVDs, one can target a specific ~15–20 bp sequence. Two TALENs are designed to bind opposite strands with a spacer in between; when both bind, the FokI halves dimerize and create a DSB. This one-to-one code of RVDs to DNA bases made TALEN design more straightforward than ZFNs [22].
TALENs generally offer very high targeting specificity. Because each TALE repeat recognizes a single nucleotide, a properly assembled TALEN pair can discriminate even single-base changes. There is no requirement for a PAM sequence, so TALENs have broad targetable range across the genome (useful if an ideal CRISPR site is lacking). In early comparisons, TALENs showed better specificity and equal or greater efficiency than first-generation ZFNs [23]. In the HPV context, TALENs can be engineered to target conserved regions of HPV genomes; for example, TALENs targeting HPV16 E7 have been created and shown to cut the viral DNA and kill HPV-positive cells [24]. An important in vivo advantage is that TALENs, being proteins, can be delivered as mRNA or protein which acts transiently, potentially reducing off-target exposure compared to a continuously expressed CRISPR plasmid [25]. One preclinical study demonstrated that intravaginal delivery of HPV-targeted TALEN plasmids in a transgenic mouse model of cervical neoplasia led to regional gene editing and therapeutic effect, highlighting feasibility of local TALEN application for HPV lesions [26].
The practicality of TALENs is hampered by the labor-intensive protein engineering required. Although modular, assembling numerous repeats (each ~100 bp of DNA code) into a plasmid is non-trivial, especially since the repeats are highly similar and prone to recombination during cloning. Methods like Golden Gate assembly have simplified this, but it remains more complex than designing a new sgRNA for CRISPR [27]. The large size of TALEN genes (each TALEN is >3 kb) means delivering a TALEN pair (≈6 kb total) often requires viral vectors with large cargo capacities or non-viral methods [27]. Off-target effects for TALENs are typically lower than early CRISPR or ZFN systems, but can occur, particularly if there are multiple similar sequences (due to TALEN’s tolerance for one or two mismatches) [28]. Interestingly, increasing TALEN DNA-binding affinity (for instance, using TALEN variants with different N-terminal domains or RVDs to boost binding) can increase off-target cutting as one study showed. Thus, there is a trade-off between TALEN activity and specificity, and careful design is needed [29]. In summary, TALENs are a powerful tool with proven efficacy in HPV gene targeting, but their use has been somewhat eclipsed by CRISPR’s ease of use. They may still hold niche value, especially where CRISPR off-target concerns, or PAM site absences make a protein-based approach advantageous.

3.3. ZFNs

Zinc-finger nucleases were the first widely adopted engineered nucleases, consisting of a DNA-binding domain made of C2H2 zinc-finger modules linked to a FokI nuclease domain. Each zinc-finger protein (ZFP) module typically recognizes a 3–4 bp DNA sequence; by concatenating 3–6 fingers, ZFNs can be designed to bind ~9–18 bp per monomer. [30]. Like TALENs, ZFNs function as dimers that cut when two FokI domains dimerize on adjacent target sites [31]. Custom ZFN design is complex: the DNA recognition of each finger can be influenced by its neighbors, and engineering a new ZFN often required expert or selection-based methods to identify fingers that bind the desired sequence with high fidelity [32]. Despite these challenges, ZFNs paved the way for clinical gene editing—for example, a ZFN therapy to disrupt the CCR5 gene in T-cells was the first gene-editing approach tested in humans (for HIV infection) and showed safety and some efficacy [33].
ZFNs are smaller in gene size than TALENs or Cas9, which is useful for delivery constraints. A pair of ZFNs can often fit into a single AAV vector (since each ZFN is roughly 1 kb, compared to a ~4 kb Cas9) [34]. Early-generation ZFNs, once successfully engineered, achieved effective genome editing and helped establish proof-of-concept for therapeutic gene editing. In theory, ZFNs can target any sequence (no PAM needed), though in practice engineering challenges made some sequences inaccessible. For HPV, ZFNs were less commonly reported than CRISPR/TALENs, but researchers have at-tempted to create ZFNs against HPV oncogenes [35]. One study designed ZFNs targeting HPV16/18 E7, demonstrating cleavage of the viral gene in cervical cancer cells. In combination with other therapies, such ZFNs showed potential to suppress HPV+ tumor cell growth [36].
The primary drawback of ZFNs is the difficulty and cost of custom design for new targets. Unlike TALENs (with a simpler one-repeat-per-base code) or CRISPR (guide RNA), ZFN engineering often required extensive protein optimization [30,37]. Off-target effects were a notable problem with some ZFNs; a comparative analysis using GUIDE-seq found that certain ZFNs generated hundreds of off-target cleavages throughout the genome. Interestingly, that study noted the off-target frequency of ZFNs correlated with certain amino acid compositions of the zinc-finger array (for instance, more “G” residues in the finger–DNA interface led to more off-target cuts). TALENs and CRISPR in the same comparison showed far fewer off-target events (on the order of 0–7 off-target sites) than ZFNs which in one case had ~1856 off-target sites [35]. This indicates that while ZFNs can be made very specific, some designs are quite error-prone, underscoring the need for thorough validation. In HPV research, ZFNs have taken a backseat due to these challenges. As more user-friendly tools emerged, new work on HPV tends to favor CRISPR or TALEN. Nonetheless, historical ZFN studies proved that targeting HPV genes could yield anti-tumor effects, and they provided a foundation on which the newer technologies improved.
Overall, CRISPR/Cas9 has become the predominant technology for genome editing in HPV-related research because of its programmability and efficiency. TALENs and ZFNs have been important in establishing feasibility and remain valuable where CRISPR is less ideal. In fact, a head-to-head comparison in 2021 specifically examining HPV16-targeted ZFNs, TALENs, and CRISPR/Cas9 concluded that SpCas9 was more efficient and more specific than either ZFNs or TALENs for HPV gene targeting. As shown by GUIDE-seq analysis, CRISPR/Cas9 had the fewest off-target events in HPV16 E6/E7 regions (0–4 off-target sites) compared to TALENs (1–36) and ZFNs (dozens to hundreds). The authors recommended CRISPR/Cas9 as the preferable platform for clinical development in HPV gene therapy, while noting their data could help improve ZFN/TALEN designs [35]. Table 1 compares the key features of these major genome editing platforms.

4. Therapeutic Applications of Genome Editing for HPV

Genome editing holds promise both as an antiviral strategy to eliminate HPV infection and as a targeted anti-cancer strategy to disable HPV oncogenes in tumors. Unlike conventional treatments that indirectly affect HPV (such as immune therapies) or nonspecifically kill dividing cells (chemotherapy, radiation), genome editing directly modifies the genetic drivers of HPV-related disease. Below, we discuss three major therapeutic applications: (1) eliminating or suppressing HPV infection in affected tissues, (2) targeting the E6/E7 oncogenes in established cancers, and (3) customizing these approaches for personalized medicine.

4.1. Editing the HPV to Eliminate Infection

One approach is to treat an HPV infection at its source by directly cleaving the viral DNA in infected cells. High-risk HPV genomes exist either as episomal circular DNA or integrated segments in host chromosomes [38]. In both cases, introducing DSBs in essential viral genes can incapacitate the virus. If the viral DNA is episomal, a nuclease-induced break can cause the small viral genome to be destroyed or misrepaired such that it can no longer replicate. If the viral DNA is integrated, cutting within E6/E7 can disrupt their expression and also potentially cause the fragment to be excised from the host genome (if two cuts are made flanking the integration site) [39]. Genome editing thus offers a radical possibility: curing an HPV infection by eradicating the viral genome from the patient’s cells. This is conceptually similar to efforts using CRISPR to eliminate latent HIV or HBV infections [40], and indeed HPV shares the challenge that current antivirals cannot purge latent viral genomes.
Preclinical studies have shown promising results in using CRISPR to eliminate HPV. In vitro, CRISPR/Cas9 targeting HPV16 E6/E7 and HPV16/18 E6 selectively eliminated HPV-positive cells, while sparing HPV-negative cells [41,42]. Moreover, CRISPR targeting HPV16 E7 had no effect on HPV-negative or even HPV18-positive cells (HeLa), demonstrating genotype-specific action [43]. In an animal model of cervical HPV infection (K14-HPV16 mice), topical application of a CRISPR/Cas9 vector led to local HPV16 gene disruptions and elimination of precancerous lesions [43].
Several strategies can enhance viral genome clearance. Using dual sgRNAs to cut out a large segment of the HPV genome (such as the entire E6/E7 region) can cause the fragment to be deleted and degraded, as demonstrated with HPV18 where dual CRISPR cuts deleted E6/E7 with high efficiency [22]. Base-editing approaches could also be used to introduce stop codons or inactivate key viral genes without DSBs, which might mitigate concerns about chromosomal translocations in integrated HPV. Although still experimental, the adaptability of CRISPR systems means base editors or prime editors could be repurposed to target HPV sequences [44]. Another possibility is prophylactic gene editing. In high-risk populations (e.g., immunosuppressed individuals), gene editing might be used to prevent progression from persistent infection to neoplasia [45].
Delivery remains a major challenge, particularly for infections in the anogenital tract. Non-invasive methods such as nanoparticle-encapsulated Cas9 RNPs or lentiviral sprays are being developed [46,47]. One study used a pH-sensitive cationic liposome to deliver Cas9 mRNA and sgRNAs to cervical cancer cells, achieving ~70% editing efficiency of HPV16 E7 in vitro [48]. Such formulations could potentially be applied to an infected epithelium in vivo to produce localized editing. Indeed, CRISPR as an antiviral is being tested for other viruses (e.g., a clinical trial uses CRISPR to excise HIV proviral DNA ), highlighting a growing precedent for this strategy [49].

4.2. Targeting HPV Oncogenes (E6 Aand E7)

HPV-driven cancers are dependent on continuous expression of E6 and E7 oncoproteins [50]. These viral proteins disable p53 and Rb, and their silencing leads to reactivation of tumor suppressor pathways and subsequent cell cycle arrest or apoptosis [51,52,53,54]. Genome editing enables specific knockout of these oncogenes, selectively killing HPV-positive cancer cells while sparing HPV-negative cells [43,53,55].
Multiple proof-of-concept studies have confirmed that editing E6/E7 has potent anti-tumor effects. For instance, using CRISPR/Cas9 to disrupt HPV16 E7 in cervical cancer cells led to upregulation of pRB and sharp inhibition of cell growth [56]. Similarly, CRISPR editing of HPV16 E6 caused apoptosis via reactivation of p53 pathways [41]. Hu et al. showed that a single sgRNA targeting HPV16 E7 reduced E7 expression and restored pRB-mediated cell cycle control [14]. In another study, multiplexed sgRNAS targeting HPV18 E6/E7 caused robust reactivation of p53 and pRB, leading to cancer cell death [22].
In vivo studies corroborate these findings. Jubair et al. systemically delivered CRISPR/Cas9 in HPV16-positive tumor-bearing mice and achieved tumor regression [52].
Treated tumors showed apoptosis as confirmed by TUNEL staining, indicating that loss of E6/E7 triggered cell death. Importantly, that study used PEGylated lipid nanoparticles to deliver the CRISPR-Cas9 plasmid to tumors, an approach that could be translatable to humans as a form of gene therapy infusion. Direct intratumoral injection of CRISPR or TALENs has also been effective in preclinical HPV models [48].
E6/E7 are ideal gene-editing targets. Unlike conventional drugs, genome editing can break non-enzymatic targets like E6/E7, which are otherwise undruggable due to their protein–protein interaction mechanisms [57]. This approach has the potential to overcome the limitations of current therapies, which do not directly eliminate viral oncogenes.
Preclinical data support a model where a CRISPR/Cas9 vector targeting HPV oncogenes is delivered into a cervical, oropharyngeal or anal tumor. Cells harboring E6/E7 are edited, leading to apoptosis and tumor shrinkage [14,43,56]. In SiHa xenograft models, intratumoral CRISPR delivery significantly inhibited tumor growth [22]. In HPV16-transgenic mice, topical application of CRISPR regressed lesions and restored normal epithelial histology [55]. These dramatic results underscore the therapeutic potential of attacking the HPV oncogene “Achilles’ heel” of these cancers. Figure 2 shows different genome editing platforms used to target HPV E6/E7.

4.3. Potential for Personalized Medicine

Personalized medicine entails tailoring treatment to the individual characteristics of each patient’s disease. Genome editing therapies for HPV can be personalized on several fronts. First, they can be customized to the patient’s HPV genotype. There are many high-risk HPV types (16, 18, 31, 33, etc.), each with slightly different DNA sequences for E6 and E7. A “one-size” CRISPR will not fit all—for example, a guide RNA targeting HPV16 E6 or E7 will not bind HPV18 oncoproteins due to sequence mismatches [53]. However, it is straightforward to sequence a patient’s HPV and then design an sgRNA (or a set of sgRNAs) that specifically target that genotype’s oncogenes. This means a CRISPR therapy could be crafted to precisely match the patient’s virus, maximizing efficacy and minimizing off-target issues. The high specificity of CRISPR is illustrated by the earlier example: a guide for HPV16 had no effect on HPV18-positive cells, so treatment must be genotype-specific. In practice, one could have a panel of pre-validated guides for the most common HPV types. After testing the patient’s biopsy and confirming, say, HPV33 is present, the clinician would choose the HPV33-specific CRISPR vector for therapy.
Second, beyond HPV type, personalized editing might consider the patient’s tumor genetics. Some HPV-associated cancers accumulate additional mutations (in host genes) that could influence response. It is conceivable to edit not just HPV genes but also certain host genes to enhance treatment. For instance, if a tumor has a particular activated oncogene that is drug-resistant, an adjunct CRISPR could knock that out too. While primarily speculative, CRISPR’s multiplexing ability allows simultaneously targeting multiple DNA sites. In a personalized context, one could target HPV E6/E7 and a host checkpoint gene in the same treatment, tailoring to that tumor’s unique profile.
Another aspect is using gene editing to engineer personalized immune cells. Although not direct editing of HPV genes, it is worth noting that CRISPR is being used to create personalized cell therapies (e.g., editing a patient’s T cells to enhance their cancer-fighting ability ) [58]. For HPV-related cancers, one might extract T cells from the patient, use CRISPR to insert a T-cell receptor that recognizes an HPV E6 peptide (or knock out inhibitory receptors), and reinfuse them—a personalized adoptive cell therapy approach. In fact, a recent trial used T cells engineered with an HPV16 E6-specific T-cell receptor in patients with cervical cancer, and a subset of patients saw tumor regression [59]. Those T cells were not edited by CRISPR (they were transduced), but future iterations could use CRISPR to improve their safety (e.g., knock out native TCR or PD-1). This blurs into immunotherapy, but it highlights how gene editing can personalize treatment: either by directly editing the tumor’s vulnerabilities or by tailoring the patient’s immune weaponry.
Lastly, personalized medicine is about precision—treating the disease with minimal collateral damage. Genome editing’s precision aligns well with this goal. With improved target specificity, a CRISPR treatment for HPV could be extremely precise (hitting only cells with the virus). Even among those cells, if, say, a patient had a rare single-nucleotide polymorphism in the target region that could block guide binding, the treatment could be adjusted (a different guide or base editor chosen). Such fine-tuning is not possible with conventional drugs. In summary, genome editing therapies for HPV can be individualized by HPV type and potentially by patient-specific factors, embodying the personalized medicine paradigm of delivering the right treatment to the right patient at the right genetic target. Figure 3 shows some of the personalized genome editing strategies like genome-specific guide RNA creation, multiplexed CRISPR and targeting patient-specific polymorphisms.

5. Preclinical and Clinical Research Progress

5.1. Preclinical Efficacy in Models

Preclinical studies have laid the groundwork for genome editing therapies targeting HPV, providing compelling evidence that disruption of viral oncogenes can suppress tumor growth and induce apoptosis in HPV-positive cells. Zhen et al. demonstrated that CRISPR/Cas9-mediated targeting of HPV16 E6 and E7 led to suppression of tumor growth in both in vitro and in vivo cervical cancer models, validating the approach as a promising therapeutic strategy [41]. Similarly, Hu et al. employed TALENs in HPV-transgenic mouse models, resulting in lesion regression, underscoring the therapeutic utility of earlier genome editing platforms [14].
Subsequent advancements introduced dual-targeting strategies, using paired sgRNAs to simultaneously disrupt both E6 and E7, preventing the compensatory survival effect that might occur if only one oncogene is inactivated. Jubair et al. applied this dual-targeting approach in a murine cervical cancer model and achieved complete tumor regression. Mechanistic analysis revealed increased expression of apoptotic markers, confirming oncoprotein inactivation as the principal mechanism of action [52].
In addition to efficacy, delivery and safety have been central concerns in preclinical models. Various delivery vehicles have been explored, including plasmid vectors, mRNA, viral systems, and ribonucleoprotein (RNP) complexes. Notably, nanoparticle-based delivery of Cas9 mRNA targeting HPV E6/E7 significantly suppressed tumor growth in murine models when combined with docetaxel [60]. These formulations hold promise for systemic administration, akin to traditional chemotherapeutics, and may eventually enable broader application in metastatic settings.
Off-target effects remain a key safety consideration. Cui et al. compared zinc finger nucleases (ZFNs), TALENs, and CRISPR-Cas9 targeting HPV16 sequences using genome-wide unbiased identification of DSBs enabled by sequencing (GUIDE-seq). Their results showed CRISPR-Cas9 exhibited the most favorable specificity profile, with minimal off-target activity in E6/E7 regions [35]. This finding supports the continued refinement of sgRNA designs and highlights the importance of specificity profiling in translational pipelines.
Beyond cervical cancer, genome editing is being explored in other HPV-related malignancies, including head and neck squamous cell carcinoma (HNSCC). While delivery to deep-seated oropharyngeal tumors presents logistical challenges, early studies suggest viral vector-mediated delivery, such as oncolytic adenoviruses carrying CRISPR-Cas9, may be feasible [61]. Moreover, patient-derived xenograft (PDX) models from HPV-positive cancers are increasingly used to assess the efficacy and safety of genome editing approaches, providing a platform for preclinical validation in anatomically and genetically relevant systems [62]. Table 2 and Table 3 summarize the in vitro & in vivo representative preclinical studies utilizing these genome editing technologies against HPV, demonstrating the progression from early ZFN/TALEN attempts to recent CRISPR successes.

5.2. Clinical Trials and Studies

Clinical translation of HPV-targeted genome editing is in its infancy, with only a limited number of trials currently underway. The first registered clinical trial (NCT03057912) evaluating gene editing for HPV was initiated by Sun Yat-sen University in China. This open-label Phase I study aims to assess the safety and efficacy of CRISPR/Cas9 or TALEN plasmids targeting E6/E7, administered intravaginally in women with persistent HPV16 or HPV18 infection and cervical intraepithelial neoplasia (CIN) grade I [73]. While the trial status is currently “unknown,” its initiation marks a milestone in the clinical application of CRISPR-based antivirals for oncogenic viruses.
The design of this trial capitalizes on the localized and accessible nature of CIN lesions, providing a low-risk environment for first-in-human evaluation. The gene therapy is delivered topically, minimizing systemic exposure, and the primary endpoints include lesion regression and HPV clearance. If successful, this study would provide the first human evidence of CRISPR-mediated viral genome eradication, supporting a new class of antiviral therapeutics.
Other clinical efforts involve gene-edited immune cells in HPV-positive cancers. For instance, a trial in China (NCT02793856) employed CRISPR to knock out the PD-1 gene in autologous T cells, which were then reinfused into patients with HPV-positive metastatic non-small cell lung cancer. The trial reported no serious adverse events linked to gene editing, establishing preliminary safety of CRISPR-engineered cell therapies in cancer patients [74].
Additionally, immunotherapies engineered to recognize HPV antigens have entered clinical development. A notable example includes the use of T cells engineered with an HPV16 E6-specific T-cell receptor (TCR) in patients with metastatic cervical cancer. In a Phase I/II trial, 2 of 12 patients demonstrated objective responses [59]. Although these cells were transduced rather than edited, future iterations are expected to incorporate CRISPR for precise engineering—such as endogenous TCR knockout to prevent mispairing or PD-1 deletion to enhance persistence.
While no genome editing therapies for HPV have received regulatory approval to date, analogous advances in other viral targets provide important precedence. Trials targeting integrated HIV DNA with CRISPR (e.g., EBT-101 for excision of latent HIV) have entered clinical stages [49], suggesting that similar strategies could be applicable to persistent HPV infection. The success of such trials will inform delivery platforms, safety assessments, and regulatory pathways for HPV-directed editing.
Moreover, therapeutic vaccines such as VGX-3100 (targeting HPV16/18 E6/E7 DNA) have demonstrated efficacy in CIN2/3, VIN2/3, AIN2/3 and PAIN2/3 regression, providing a clinical benchmark [75,76,77,78,79]. In a Phase II trial, VGX-3100 achieved 49.5% histopathologic regression in cervical HSIL versus 30.6% with placebo [76]. Therefore, any CRISPR-based HPV therapy must match or exceed these efficacy rates and safety profiles to justify clinical deployment. Table 4 gives an overview of the ongoing and completed clinical trials utilizing gene editing tools to combat HPV-associated malignancies.
In summary, the preclinical-to-clinical pipeline for genome editing in HPV-associated disease is progressing steadily. While the field is in early stages, the biological rationale, preclinical data, and emerging trial activity suggest a promising future. Continued efforts in optimizing delivery, guide design, and patient stratification will be critical for clinical success.

6. Oncological Perspectives and Integration into Treatment

Genome editing offers a novel therapeutic strategy for HPV-related diseases by integrating directly into the oncology care paradigm. Its unique mechanism—disrupting viral oncogenes—complements or potentially surpasses current therapeutic modalities including surgery, radiotherapy, chemotherapy, immunotherapy, and therapeutic vaccines [80]. Strategic deployment of gene editing must consider the disease stage, anatomical accessibility, and patient-specific factors to optimize benefit [44].

6.1. Complement to Existing Therapies

A promising application of genome editing lies in intercepting premalignant HPV-related lesions. For patients with persistent or multifocal cervical intraepithelial neoplasia (CIN), CRISPR-based therapy could offer an alternative to repeated surgical excisions, which are associated with cervical insufficiency and adverse pregnancy outcomes [81]. In particular, locally applied CRISPR therapeutics (e.g., a gel-based formulation) targeting E6/E7 oncogenes could eradicate infection and dysplastic cells while preserving cervical integrity [82].
For CIN1 cases where lesions often regress spontaneously, gene editing may facilitate lesion clearance in persistent cases, reducing the psychosocial burden associated with HPV diagnosis. In this setting, genome editing would function as a preventive or interceptive intervention, rather than replacing standard cancer therapies.
In invasive cervical or head and neck cancers, genome editing may function as an adjunct to standard therapies. Preclinical studies suggest that inactivating HPV oncogenes restores p53/Rb pathways, sensitizing tumors to radiation and chemotherapy [60,67,83]. For example, pre-treatment with E6/E7-targeted CRISPR may enhance tumor cell susceptibility to DNA damage-induced apoptosis. Additionally, adjuvant delivery of gene editing agents post-surgery could target microscopic residual HPV-positive cells, reducing recurrence risk without systemic toxicity.

6.2. Comparison with Immunotherapy

Checkpoint inhibitors such as pembrolizumab have modest efficacy in HPV-positive cancers, with objective response rates of approximately 14% in cervical cancer [84]. Adoptive T-cell therapies, including tumor-infiltrating lymphocytes (TILs) or engineered T cells expressing HPV-specific TCRs, have shown early promise but are dependent on patient immune status [85]. In contrast, genome editing directly induces cytotoxicity in HPV-positive cells and may be effective in immunocompromised individuals, such as transplant recipients or HIV-positive patients with HPV-associated lesions [80].
Importantly, genome editing may also synergize with immunotherapies. The immunogenic cell death following CRISPR-mediated E6/E7 knockout can release viral antigens and prime an anti-tumor immune response. This “in situ vaccination” effect may enhance the efficacy of checkpoint inhibitors [86]. Furthermore, gene editing-induced tumor cell death may shift the tumor microenvironment towards an inflamed, immuno-permissive state, improving immunotherapy outcomes [87].

6.3. Versus Chemoradiation

Chemoradiation remains the standard of care for locally advanced cervical cancer [88]. However, recurrence rates remain significant in patients with bulky or high-risk tumors. Genome editing offers a tumor-specific alternative, potentially avoiding systemic side effects such as nephrotoxicity, neurotoxicity, nausea, and infertility associated with cisplatin-based regimens [89].
Compared to radiotherapy, gene editing may induce less off-target damage to adjacent normal tissues, provided precise delivery is achieved. This could preserve organ function and reduce long-term sequelae such as fibrosis, vaginal stenosis, and bowel injury [90]. However, challenges remain in ensuring that gene editing agents reach the same breadth of tumor burden as ionizing radiation, especially in diffusely infiltrative disease.

6.4. Unique Benefit—Viral Specificity

One perspective often voiced in oncology is that virally induced tumors present unique targets (the viral genes) that non-virally induced tumors lack. This has been exploited by therapeutic vaccines and by T-cell therapies [91]. Genome editing is another way to exploit the virus-specific target. It is akin to a magic bullet that only cancer cells (with the virus) will absorb. This is particularly relevant in multifocal disease—for example, HPV can cause field cancerization in the urogenital tract; after one lesion is treated, new ones can appear [92]. A systemic or regional gene editing treatment might simultaneously treat all lesions, something surgery or localized radiation cannot do easily. In patients with HPV-driven oropharyngeal cancer, often multiple tumors or extensive carcinoma in situ can be present (like spreading along the mucosa) [93]; gene editing delivered by a viral vector that diffuses through the mucosa could potentially treat a broad area.

6.5. Safety Considerations in Oncology

Oncologists will be cautious about gene editing toxicity. Off-target cuts in the human genome that hit, say, a tumor suppressor or essential gene in normal cells could theoretically initiate secondary malignancies or other issues. Long-term follow-up will be needed for any gene therapy to ensure we are not causing new problems while curing HPV. That said, traditional treatments like radiation also cause DNA damage and carry a risk of secondary cancers; the hope is a targeted gene editor will actually be safer in that regard by focusing the breaks where you intend them [94]. Preclinical data so far, as mentioned, show minimal off-target effects with properly designed CRISPR in the HPV context, which is encouraging.

6.6. Patient Acceptance and Feasibility

From a patient-centered perspective, gene editing could be appealing as a non-invasive or minimally invasive outpatient therapy. Cervical injections, topical gels, or intravenous infusions of nanoformulated CRISPR may reduce treatment burdens compared to hospital-based radiation or surgical procedures [95]. Moreover, reduced systemic toxicity could improve quality of life during therapy.
Nonetheless, public perception of gene editing remains a potential barrier. Misconceptions about germline modification or irreversible DNA damage may evoke fear or resistance. Effective communication strategies must emphasize the somatic and therapeutic nature of the intervention, and the virus-specific targeting of cancer cells.

7. Challenges and Future Prospects

While the promise of genome editing for HPV is great, several challenges must be overcome before these therapies become routine.

7.1. Technical Hurdles

The foremost technical challenge is delivery. Getting the genome editing molecules to the right cells in the patient—and enough of those cells—is non-trivial [96]. For cervical lesions, direct application is possible (e.g., using a cervical spray or injection), but for internal tumors like oropharyngeal cancer or metastatic disease, one might need systemic delivery [97,98]. Viral vectors (such as adenovirus or AAV) can deliver genes efficiently, but they come with size limits and potential immune reactions [99]. Non-viral methods like lipid nanoparticles or polymer nanoparticles are being actively developed; Poly (β-Amino Ester)-based nanoparticles were used successfully to deliver CRISPR to HPV tumors [100,101,102]. Optimizing these carriers for human use is ongoing—for example, designing pH-responsive or tissue-targeted nanoparticles to improve uptake by cervical or tumor tissues [48].

7.2. Efficiency of Editing

In a tumor, if only 50% of cells get edited, the remaining 50% might continue to proliferate (though they might have less growth advantage if their neighbors are dying and releasing cytokines). A high editing rate will be important for robust efficacy. Strategies like using base editors or Cas nucleases fused to cytidine deaminase (which can induce mutations without needing a cut on every allele) could help amplify the effect even if delivery is not 100% [103].
Off-target effects and genomic safety remain significant technical and scientific challenges. Although improved guide design has reduced off-target cutting, we cannot assume zero off-targets. Each new guide intended for clinical use will require comprehensive screening (perhaps with unbiased methods like GUIDE-seq or DISCOVER-seq in relevant human cell types) to ensure it does not significantly hit other genomic sites. It is reassuring that well-designed guides for 3 HPV16 critical target genes in one study showed very minimal detectable off-target activity [35]. Additionally, since the target cells (e.g., cervical epithelium) can be biopsied, there is potential to verify editing events after treatment to ensure only the desired edits occurred. If undesired mutations are found, it would raise flags. Another nuance is if the HPV genome is integrated in a critical location in the host genome—cutting it out might cause a large deletion in a host chromosome. For example, if HPV inserted within a tumor suppressor gene, using CRISPR to cut out HPV might also excise part of that suppressor. This scenario is unpredictable and rare, but possible. It might be mitigated by understanding common integration sites (HPV often integrates in fragile sites or in certain loci; if one knows the integration, one could plan the cut strategy accordingly).

7.3. Immune Responses and Safety

Delivering a bacterial protein like Cas9 or foreign TALE proteins could provoke an immune response [104]. This is double-edged: on one hand, a localized immune response might help kill tumor cells (good for efficacy), but on the other hand it could limit repeated dosing or cause inflammatory side effects. Immune responses against AAV or adenoviral vectors are well-documented, and repeated dosing can be problematic if neutralizing antibodies form [105]. For a one-time therapy in a localized site, this might be acceptable. Using human protein-based editors (e.g., a human-derived endonuclease, though none are in clinical use yet) is an interesting idea but not currently practical. Immune suppression during treatment is another approach (e.g., giving a short course of steroids or immunosuppressants to allow the editor to do its job without being cleared too fast). The balance between harnessing inflammation to attack the virus-infected cells and avoiding systemic autoimmunity will need careful management.

7.4. Ethical and Regulatory Concerns

Ethically, somatic genome editing for life-threatening conditions is generally accepted, but caution is paramount. It is crucial to emphasize that these interventions would not affect germline DNA—especially important for therapies in the reproductive tract like cervical treatments, to reassure that we are not altering eggs or sperm. In preclinical models, CRISPR delivered to the cervix did not result in detectable edits in the ovaries or distant tissues, suggesting localization can be achieved. Regulators will require extensive safety data, likely including animal studies in non-human primates or other models, to show that the gene therapy does not cause unintended consequences. Long-term follow-up of trial participants (for many years) will be mandated to monitor for any delayed adverse effects like secondary cancers. Another ethical aspect is informed consent and patient understanding—“gene editing” might sound frightening; clear education is needed so patients understand the therapy’s intent and risks.
Cost and access are also considerations for the future. Gene therapies tend to be expensive. If a CRISPR-based HPV cure costs significantly more than, say, surgery or standard chemo, that could limit adoption, especially in low-resource settings where HPV disease is most prevalent. Simplifying the treatment (maybe one or two outpatient applications) and scaling manufacturing could help reduce cost. The good news is that plasmids or RNAs are relatively inexpensive to produce compared to monoclonal antibodies or cell therapies. If the field can produce “off-the-shelf” CRISPR kits for common HPV types, it might be cost-effective in the long run, especially considering the healthcare savings from preventing cancers.

7.5. Future Research Directions

Future work will likely explore refined genome editors—such as base editors (which can introduce stop codons in E6/E7 without a DSB) or prime editors (which could precisely insert disruptive sequences or corrections) [44]. These next-generation tools might avoid DSB-associated risks like chromosomal rearrangements. Base editing of HPV, for instance, could mutate the start codon of E7 or create nonsense mutations in E6, permanently inactivating them without cutting the DNA—an elegant solution if it can be delivered efficiently. Already, researchers have noted that given the small size of HPV genes, even a single base change can knock out oncoprotein function, so base editors could be very effective.
Another area is expanding targetable sites. Cas9 requires a PAM (NGG), which might not be optimally placed to cut some HPV genes. Using other nucleases like Cas12a/Cpf1 (which has a different PAM and staggered cut) could open up more targeting options [106]. Indeed, one study reported using CRISPR/Cas12a to detect HPV DNA [107]. The same enzyme could be used to cut HPV DNA with a TTTV PAM requirement. Additionally, the concept of repurposing CRISPR as a suppressor (CRISPRi) could be explored: instead of cutting, a dCas9 fused to a repressor could sit on the HPV promoter region and shut off E6/E7 transcription [108]. This might be safer (no breaks at all), but would require continuous presence or expression, which is a different therapeutic model (more akin to a drug you have to keep taking, rather than a one-and-done edit).

7.6. Translational Potential

The next few years will be telling as initial clinical data emerges. If the early CIN trial shows positive results, we might see rapid moves to test such therapies in high-grade lesions (CIN3) or even as an adjunct in early cervical cancers to preserve fertility (e.g., treating with CRISPR instead of doing a radical trachelectomy for a small tumor). For head and neck cancers, perhaps a local injection during endoscopy could be trialed. There is also interest in HPV-associated anal intraepithelial neoplasia (AIN) in high-risk groups (like HIV-positive individuals)—a gene editing approach could be very useful there since those lesions are often multifocal and recurrent.
Regulatory approval will depend on demonstrating that the benefits (cancer prevention or treatment) outweigh the risks of off-target effects. Given that current treatments for advanced HPV cancers still have significant failure rates, a successful new modality would be welcome. We might imagine a future where persistent HPV infection is treated akin to how we treat Helicobacter pylori to prevent stomach cancer—i.e., you diagnose an HPV infection that is not clearing, and instead of just monitoring, you give gene therapy to knock it out [109].

8. Discussion

Genome editing technologies have generated substantial enthusiasm for treating HPV-driven cancers because they can directly disrupt viral oncogenes and restore tumor-suppressor pathways, with consistent proof-of-concept across in vitro and in vivo models [14,22,23,24,41,52,55,62,110]. The technological toolbox continues to expand with base and prime editing, which may improve targeting scope and precision, and there is growing interest in combining editing with immuno-oncology to enhance anti-tumor responses [44,58,72,84,85,103].
Despite these opportunities, translation requires a clear view of the formidable challenges ahead. First, efficient delivery to HPV-infected epithelia and solid tumors remains a central barrier. While vectors and nanocarriers show promise—including adenoviral/AAV and lentiviral platforms, as well as lipid and polymer systems—achieving safe, tissue-specific, and scalable delivery is unresolved [32,95,96,99,100,102,104,105]. Practical routes (e.g., intravaginal or local mucosal administration) are being explored to improve exposure at the disease site, but clinical maturation is still early [48,82,100,101,102].
Second, off-target activity demands careful risk management, particularly given HPV’s frequent integration into host genomes. Unintended editing near integration sites could disrupt tumor suppressors or activate oncogenes. Although the probability of HPV integrating directly into a tumor suppressor is relatively low, integration events are often non-random and tend to occur near fragile genomic regions or sites of instability. In many cases, integration may already compromise tumor suppressor activity, yet excision via genome editing at such loci still carries the risk of collateral damage if essential host genes are affected [20,28,35,103,106,111]. Comparative and genome-wide profiling studies underscore that platform choice, guide design, and PAM-proximal/distal sequence context all shape specificity, yet risk elimination is not guaranteed [28,35,94,97]. Continued use of high-fidelity nucleases, rigorous in silico/off-target prediction, and orthogonal validation will be essential before broad clinical uptake [28,35,103,106].
Third, cost and accessibility must also be considered. The global burden of HPV-driven cancers is greatest in low- and middle-income countries (LMICs), where the infrastructure for advanced molecular therapeutics is least available [2,12,18]. Without deliberate strategies to address affordability and implementation, genome editing may inadvertently widen existing disparities in cancer care. In this context, potential solutions include international collaborations, tiered pricing strategies, and public–private partnerships to subsidize access in high-burden regions. Incorporating such funding models into early planning will be critical to ensure that these therapies, if proven effective, are not restricted to high-resource settings alone [18,99].
Finally, ethical and governance issues require sustained attention. Although reproductive or germline applications are not the focus here, broader debates around consent, long-term follow-up, and societal acceptability remain directly relevant to clinical translation of somatic editing technologies [99,111]. Transparent regulation, community engagement, and long-term safety monitoring will be pivotal to public trust.
Crucially, the field should draw lessons from past gene-therapy attempts, where progress was hampered by delivery inefficiency, vector immunogenicity, and safety signals that limited durability and scalability [99,105]. Unless these historical obstacles are addressed head-on—with better vectors, improved immune evasion, and stringent safety analytics—HPV-targeted genome editing may face similar headwinds.
Taken together, a balanced perspective is warranted: genome editing against HPV offers a compelling mechanistic rationale and robust preclinical activity, yet its clinical success will depend on solving delivery and specificity, ensuring equitable access, and maintaining rigorous ethical oversight. In this light, editing should be pursued as a complement, rather than a replacement for existing prevention and treatment frameworks, while these barriers are systematically reduced [72,84,85].

9. Conclusions

Genome editing represents a promising avenue for addressing HPV-driven cancers by directly targeting viral oncogenes and restoring host tumor suppressor pathways. Preclinical studies provide compelling proof-of-concept, and ongoing innovations such as base and prime editing expand the potential applications. Nevertheless, significant barriers remain, including delivery inefficiency, off-target risks in the context of viral integration, ethical considerations, and challenges of cost and accessibility in low-resource settings. Addressing these obstacles will be essential before clinical translation can be achieved. Rather than a replacement for existing strategies, genome editing should be viewed as a potential complement to vaccination, screening, and conventional therapies, with future progress depending on rigorous preclinical validation, responsible governance, and equitable access.

Author Contributions

Conceptualization, M.O.C., M.S., B.Y., M.O. and G.H.A.; methodology, M.O.C., M.S., B.Y., M.O. and G.H.A.; software, M.O.C., M.S., B.Y., M.O. and G.H.A.; validation, M.O.C., M.S., B.Y., M.O. and G.H.A.; formal analysis, M.O.C., M.S., B.Y., M.O. and G.H.A.; investigation, M.O.C., M.S., B.Y., M.O. and G.H.A.; resources, M.O.C., M.S., B.Y., M.O. and G.H.A.; data curation, M.O.C., M.S., B.Y., M.O. and G.H.A.; writing—original draft preparation, M.O.C., M.S., B.Y., M.O. and G.H.A.; writing—review and editing, M.O.C., M.S., B.Y., M.O. and G.H.A.; visualization, M.O.C., M.S., B.Y., M.O. and G.H.A.; supervision, M.O.C., B.Y., M.O. and G.H.A.; project administration, M.O.C., B.Y., M.O. and G.H.A.; funding acquisition, B.Y., M.O. and G.H.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Stanley, M. Pathology and epidemiology of HPV infection in females. Gynecol. Oncol. 2010, 117 (Suppl. S2), S5–S10. [Google Scholar] [CrossRef]
  2. 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]
  3. Hausen, H. Papillomaviruses and cancer: From basic studies to clinical application. Nat. Rev. Cancer 2002, 2, 342–350. [Google Scholar] [CrossRef]
  4. Moody, C.A.; Laimins, L.A. Human papillomavirus oncoproteins: Pathways to transformation. Nat. Rev. Cancer 2010, 10, 550–560. [Google Scholar] [CrossRef]
  5. McLaughlin-Drubin, M.E.; Münger, K. The human papillomavirus E7 oncoprotein. Virology 2009, 384, 335–344. [Google Scholar] [CrossRef] [PubMed]
  6. Münger, K.; Howley, P.M. Human papillomavirus immortalization and transformation functions. Virus Res. 2002, 89, 213–228. [Google Scholar] [CrossRef] [PubMed]
  7. Doorbar, J. Molecular biology of human papillomavirus infection and cervical cancer. Clin. Sci. 2006, 110, 525–541. [Google Scholar] [CrossRef] [PubMed]
  8. Koutsky, L. Epidemiology of genital human papillomavirus infection. Am. J. Med. 1997, 102, 3–8. [Google Scholar] [CrossRef]
  9. Arbyn, M.; Castellsagué, X.; de Sanjosé, S.; Bruni, L.; Saraiya, M.; Bray, F.; Ferlay, J. Worldwide burden of cervical cancer in 2008. Ann. Oncol. 2011, 22, 2675–2686. [Google Scholar] [CrossRef]
  10. The FUTURE II Study Group. Quadrivalent vaccine against HPV to prevent high-grade cervical lesions. N. Engl. J. Med. 2007, 356, 1928–1943. [Google Scholar] [CrossRef]
  11. Drolet, M.; Bénard, É.; Pérez, N.; Brisson, M.; HPV Vaccination Impact Study Group. Population-level impact and herd effects following the introduction of human papillomavirus vaccination programmes: Updated systematic review and meta-analysis. Lancet 2019, 394, 497–509. [Google Scholar] [CrossRef] [PubMed]
  12. Pimple, S.A.; Mishra, G.A. Global strategies for cervical cancer prevention and screening. Minerva Ginecol. 2019, 71, 313–320. [Google Scholar] [CrossRef] [PubMed]
  13. Jemal, A.; Center, M.M.; DeSantis, C.; Ward, E.M. Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol. Biomarkers Prev. 2010, 19, 1893–1907. [Google Scholar] [CrossRef]
  14. Hu, Z.; Yu, L.; Zhu, D.; Ding, W.; Wang, X.; Zhang, C.; Wang, L.; Jiang, X.; Shen, H.; He, D.; et al. Disruption of HPV16-E7 by CRISPR/Cas System Induces Apoptosis and Growth Inhibition in HPV16 Positive Human Cervical Cancer Cells. BioMed Res. Int. 2014, 1, 612823. [Google Scholar] [CrossRef] [PubMed]
  15. Ravanlo, Z.Z.; Gholami, S.; Baghi, H.B. HPV-Specific Antivirals: Disarming Viral Entry and Disruption of Replication. Microbe 2025, 9, 100593. [Google Scholar] [CrossRef]
  16. Egawa, N.; Doorbar, J. The low-risk papillomaviruses. Virus Res. 2017, 231, 119–127. [Google Scholar] [CrossRef]
  17. Ntuli, L.; Mtshali, A.; Mzobe, G.; Liebenberg, L.J.P.; Ngcapu, S. Role of Immunity and Vaginal Microbiome in Clearance and Persistence of Human Papillomavirus Infection. Front. Cell. Infect. Microbiol. 2022, 12, 927131. [Google Scholar] [CrossRef]
  18. Cuzick, J.; Adcock, R.; Kinney, W.; Castle, P.E.; Robertson, M.; McDonald, R.M.; Stoler, M.H.; Du, R.; Wheeler, C.M. Impact of HPV testing in Opportunistic Cervical Screening: Support for Primary HPV Screening in the United States. Int. J. Cancer 2023, 153, 83. [Google Scholar] [CrossRef]
  19. Joung, J.K.; Sander, J.D. TALENs: A widely applicable technology for genome editing. Nat. Rev. Mol. Cell. Biol. 2013, 14, 49–55. [Google Scholar] [CrossRef]
  20. Sander, J.D.; Joung, J.K. CRISPR–Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 2014, 32, 347–355. [Google Scholar] [CrossRef]
  21. Urnov, F.D.; Rebar, E.J.; Holmes, M.C.; Zhang, H.S.; Gregory, P.D. Genome editing with engineered zinc finger nucleases. Nat. Rev. Genet. 2010, 11, 636–646. [Google Scholar] [CrossRef]
  22. Ling, K.; Yang, L.; Yang, N.; Chen, M.; Wang, Y.; Liang, S.; Li, Y.; Jiang, L.; Yan, P.; Liang, Z. Gene Targeting of HPV18 E6 and E7 Synchronously by Nonviral Transfection of CRISPR/Cas9 System in Cervical Cancer. Hum. Gene Ther. 2020, 31, 297–308. [Google Scholar] [CrossRef]
  23. Khan, S.H. Genome-Editing Technologies: Concept, Pros, and Cons of Various Genome-Editing Techniques and Bioethical Concerns for Clinical Application. Mol. Ther. Nucleic Acids 2019, 16, 326–334. [Google Scholar] [CrossRef]
  24. Liu, W.; Jiang, Y.; Wang, C.; Wang, M.; Zhang, W.; Ren, H.; Xu, S.; Qin, J.; Liu, P.; Jin, L.; et al. Towards the elimination of infectious HPV: Exploiting CRISPR/Cas innovations. Front. Cell. Infect. Microbiol. 2025, 15, 1627668. [Google Scholar] [CrossRef]
  25. Popovitz, J.; Sharma, R.; Hoshyar, R.; Soo Kim, B.; Murthy, N.; Lee, K. Gene editing therapeutics based on mRNA delivery. Adv. Drug Deliv. Rev. 2023, 200, 115026. [Google Scholar] [CrossRef]
  26. Ren, C.; Li, X.; Mao, L.; Xiong, J.; Gao, C.; Shen, H.; Wang, L.; Zhu, D.; Ding, W.; Wang, H. An effective and biocompatible polyethylenimine based vaginal suppository for gene delivery. Nanomed. Nanotechnol. Biol. Med. 2019, 20, 101994. [Google Scholar] [CrossRef]
  27. Cermak, T.; Doyle, E.L.; Christian, M.; Wang, L.; Zhang, Y.; Schmidt, C.; Baller, J.A.; Somia, N.V.; Bogdanove, A.J.; Voytas, D.F. Efficient design and assembly of custom TALEN and TALE-based constructs for DNA targeting. Nucleic Acids Res. 2011, 39, e82. [Google Scholar] [CrossRef] [PubMed]
  28. Yee, J.-K. Off-target effects of engineered nucleases. FEBS J. 2016, 283, 3239–3248. [Google Scholar] [CrossRef] [PubMed]
  29. Sunami, T.; Kono, H. Balance between DNA-binding affinity and specificity enables selective recognition of longer target sequences in vivo. Protein Sci. 2019, 28, 1630–1639. [Google Scholar] [CrossRef]
  30. Davis, D.; Stokoe, D. Zinc finger nucleases as tools to understand and treat human diseases. BMC Med. 2010, 8, 42. [Google Scholar] [CrossRef] [PubMed]
  31. Cradick, T.J.; Ambrosini, G.; Iseli, C.; Bucher, P.; McCaffrey, A.P. Zfn-site searches genomes for zinc finger nuclease target sites and off-target sites. BMC Bioinform. 2011, 12, 152. [Google Scholar] [CrossRef] [PubMed]
  32. Zhu, C.; Gupta, A.; Hall, V.L.; Rayla, A.L.; Christensen, R.G.; Dake, B.; Lakshmanan, A.; Kuperwasser, C.; Stormo, G.D.; Wolfe, S.A. Using defined finger–finger interfaces as units of assembly for constructing zinc-finger nucleases. Nucleic Acids Res. 2013, 41, 2455–2465. [Google Scholar] [CrossRef] [PubMed]
  33. Tebas, P.; Stein, D.; Tang, W.W.; Frank, I.; Wang, S.Q.; Lee, G.; Spratt, S.K.; Surosky, R.T.; Giedlin, M.A.; Nichol, G.; et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N. Engl. J. Med. 2014, 370, 901–910. [Google Scholar] [CrossRef] [PubMed]
  34. Katayama, S.; Watanabe, M.; Kato, Y.; Nomura, W.; Yamamoto, T. Engineering of zinc finger nucleases through structural modeling improves genome editing efficiency in cells. Adv. Sci. 2024, 11, e2310255. [Google Scholar] [CrossRef]
  35. Cui, Z.; Liu, H.; Zhang, H.; Huang, Z.; Tian, R.; Li, L.; Fan, W.; Chen, Y.; Chen, L.; Zhang, S.; et al. The comparison of ZFNs, TALENs, and SpCas9 by GUIDE-seq in HPV-targeted gene therapy. Mol. Ther. Nucleic Acids 2021, 26, 1466–1478. [Google Scholar] [CrossRef]
  36. Ding, W.; Hu, Z.; Zhu, D.; Jiang, X.; Yu, L.; Wang, X.; Zhang, C.; Wang, L.; Ji, T.; Li, K.; et al. Zinc Finger Nucleases Targeting the Human Papillomavirus E7 Oncogene Induce E7 Disruption and a Transformed Phenotype in HPV16/18-Positive Cervical Cancer Cells. Clin. Cancer Res. 2014, 20, 6495–6503. [Google Scholar] [CrossRef]
  37. Shankar, S.; Sreekumar, A.; Prasad, D.; Das, A.V.; Pillai, M.R. Genome editing of oncogenes with ZFNs and TALENs: Caveats in nuclease design. Cancer Cell Int. 2018, 18, 169. [Google Scholar] [CrossRef]
  38. Hong, D.; Liu, J.; Hu, Y.; Lu, X.; Li, B.; Li, Y.; Hu, D.; Lu, W.; Xie, X.; Cheng, X. Viral E6 is overexpressed via high viral load in invasive cervical cancer with episomal HPV16. BMC Cancer 2017, 17, 136. [Google Scholar] [CrossRef]
  39. Bi, Y.; Sun, L.; Gao, D.; Ding, C.; Li, Z.; Li, Y.; Cun, W.; Li, Q. High-efficiency targeted editing of large viral genomes by RNA-guided nucleases. PLoS Pathog. 2014, 10, e1004090. [Google Scholar] [CrossRef]
  40. Bhowmik, R.; Chaubey, B. Crispr/cas9: A tool to eradicate HIV-1. AIDS Res. Ther. 2022, 19, 58. [Google Scholar] [CrossRef]
  41. Zhen, S.; Lu, J.-J.; Wang, L.-J.; Sun, X.-M.; Zhang, J.-Q.; Li, X.; Luo, W.-J.; Zhao, L. In Vitro and In Vivo Synergistic Therapeutic Effect of Cisplatin with Human Papillomavirus16 E6/E7 CRISPR/Cas9 on Cervical Cancer Cell Line. Transl. Oncol. 2016, 9, 498–504. [Google Scholar] [CrossRef]
  42. Ehrke-Schulz, E.; Heinemann, S.; Schulte, L.; Schiwon, M.; Ehrhardt, A. Adenoviral Vectors Armed with PAPILLOMAVIRUs Oncogene Specific CRISPR/Cas9 Kill Human-Papillomavirus-Induced Cervical Cancer Cells. Cancers 2020, 12, 1934. [Google Scholar] [CrossRef]
  43. Gao, C.; Wu, P.; Yu, L.; Liu, L.; Liu, H.; Tan, X.; Wang, L.; Huang, X.; Wang, H. The application of CRISPR/Cas9 system in cervical carcinogenesis. Cancer Gene Ther. 2022, 29, 466–474. [Google Scholar] [CrossRef] [PubMed]
  44. Xu, W.; Zhang, S.; Qin, H.; Yao, K. From bench to bedside: Cutting-edge applications of base editing and prime editing in precision medicine. J. Transl. Med. 2024, 22, 1133. [Google Scholar] [CrossRef]
  45. Haddad, C.O.; Kalt, I.; Shovman, Y.; Xia, L.; Schlesinger, Y.; Sarid, R.; Parnas, O. Targeting the kaposi’s sarcoma-associated herpesvirus genome with the crispr-cas9 platform in latently infected cells. Virol. J. 2021, 18, 56. [Google Scholar] [CrossRef]
  46. Tsuchida, C.A.; Wasko, K.M.; Hamilton, J.R.; Doudna, J.A. Targeted nonviral delivery of genome editors in vivo. Proc. Natl. Acad. Sci. USA 2024, 121, e2307796121. [Google Scholar] [CrossRef] [PubMed]
  47. Sinn, P.L.; Arias, A.C.; Brogden, K.A.; McCray, P.B., Jr. Lentivirus vector can be readministered to nasal epithelia without blocking immune responses. J. Virol. 2008, 82, 10684–10692. [Google Scholar] [CrossRef]
  48. Zhen, S.; Liu, Y.; Lu, J.; Tuo, X.; Yang, X.; Chen, H.; Chen, W.; Li, X. Human Papillomavirus Oncogene Manipulation Using Clustered Regularly Interspersed Short Palindromic Repeats/Cas9 Delivered by pH-Sensitive Cationic Liposomes. Hum. Gene Ther. 2020, 31, 309–324. [Google Scholar] [CrossRef] [PubMed]
  49. Excision BioTherapeutics. Clinical Trial EBT-101 for HIV CRISPR Treatment (NCT05144386). ClinicalTrials.gov.; 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT05144386 (accessed on 8 October 2025).
  50. Baba, S.K.; Alblooshi, S.S.E.; Yaqoob, R.; Behl, S.; Al Saleem, M.; Rakha, E.A.; Malik, F.; Singh, M.; Macha, M.A.; Akhtar, M.K.; et al. Human papilloma virus (hpv) mediated cancers: An insightful update. J. Transl. Med. 2025, 23, 483. [Google Scholar] [CrossRef]
  51. Kennedy, E.M.; Kornepati, A.V.R.; Goldstein, M.; Bogerd, H.P.; Poling, B.C.; Whisnant, A.W.; Kastan, M.B.; Cullen, B.R. Inactivation of the Human Papillomavirus E6 or E7 Gene in Cervical Carcinoma Cells by Using a Bacterial CRISPR/Cas RNA-Guided Endonuclease. J. Virol. 2014, 88, 11965–11972. [Google Scholar] [CrossRef]
  52. Jubair, L.; Fallaha, S.; McMillan, N.A.J. Systemic Delivery of CRISPR/Cas9 Targeting HPV Oncogenes Is Effective at Eliminating Established Tumors. Mol. Ther. 2019, 27, 2091–2099. [Google Scholar] [CrossRef] [PubMed]
  53. Khairkhah, N.; Bolhassani, A.; Rajaei, F.; Najafipour, R. Systemic delivery of specific and efficient CRISPR/Cas9 system targeting HPV16 oncogenes using LL-37 antimicrobial peptide in C57BL/6 mice. J. Med. Virol. 2023, 95, e28934. [Google Scholar] [CrossRef] [PubMed]
  54. Chen, Y.; Jiang, H.; Wang, T.; He, D.; Tian, R.; Cui, Z.; Tian, X.; Gao, Q.; Ma, X.; Yang, J.; et al. In vitro and in vivo growth inhibition of human cervical cancer cells via human papillomavirus E6/E7 mRNAs’ cleavage by CRISPR/Cas13a system. Antivir. Res. 2020, 178, 104794. [Google Scholar] [CrossRef]
  55. Hu, Z.; Ding, W.; Zhu, D.; Yu, L.; Jiang, X.; Wang, X.; Zhang, C.; Wang, L.; Ji, T.; Liu, D.; et al. TALEN-mediated targeting of HPV oncogenes ameliorates HPV-related cervical malignancy. J. Clin. Investig. 2015, 125, 425–436. [Google Scholar] [CrossRef]
  56. Ferreira, D.A.; McMillan, N.A.J.; Idris, A. Genetic deletion of HPV E7 oncogene effectively regresses HPV driven oral squamous carcinoma tumour growth. Biomed. Pharmacother. 2022, 155, 113782. [Google Scholar] [CrossRef]
  57. McLaughlin-Drubin, M.E.; Münger, K. Oncogenic activities of human papillomaviruses. Virus Res. 2009, 143, 195–208. [Google Scholar] [CrossRef]
  58. Tony, L.T.; Stabile, A.; Schauer, M.P.; Hudecek, M.; Weber, J. CAR-T Cell Therapy for Solid Tumors. Transfus. Med. Hemother. 2025, 52, 96–108. [Google Scholar] [CrossRef]
  59. Xinqiao Hospital of Chongqing. HPV-E6-Specific Anti-PD1 TCR-T Cells in the Treatment of HPV-Positive NHSCC or Cervical Cancer (NCT03578406). ClinicalTrials.gov.; 2019. Available online: https://clinicaltrials.gov/study/NCT03578406 (accessed on 8 October 2025).
  60. Li, X.; Guo, M.; Hou, B.; Zheng, B.; Wang, Z.; Huang, M.; Xu, Y.; Chang, J.; Wang, T. Crispr/cas9 nanoeditor of double knockout large fragments of e6 and e7 oncogenes for reversing drugs resistance in cervical cancer. J. Nanobiotechnol. 2021, 19, 231. [Google Scholar] [CrossRef]
  61. Ghanaat, M.; Goradel, N.H.; Arashkia, A.; Ebrahimi, N.; Ghorghanlu, S.; Malekshahi, Z.V.; Fattahi, E.; Negahdari, B.; Kaboosi, H. Virus against virus: Strategies for using adenovirus vectors in the treatment of HPV-induced cervical cancer. Acta Pharmacol. Sin. 2021, 42, 1981–1990. [Google Scholar] [CrossRef] [PubMed]
  62. Hsu, D.S.; Kornepati, A.V.R.; Glover, W.; Kennedy, E.M.; Cullen, B.R. Targeting HPV16 DNA using CRISPR/Cas inhibits anal cancer growth in vivo. Future Virol. 2018, 13, 475–482. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, Y.-C.; Cai, Z.-M.; Zhang, X.-J. Reprogrammed CRISPR-Cas9 Targeting the Conserved Regions of HPV6/11 E7 Genes Inhibits Proliferation and Induces Apoptosis in E7-Transformed Keratinocytes. Asian J. Androl 2016, 18, 475–479. [Google Scholar] [CrossRef]
  64. Lao, Y.-H.; Li, M.; Gao, M.A.; Shao, D.; Chi, C.-W.; Huang, D.; Chakraborty, S.; Ho, T.-C.; Jiang, W.; Wang, H.-X.; et al. HPV Oncogene Manipulation Using Nonvirally Delivered CRISPR/Cas9 or Natronobacterium gregoryi Argonaute. Adv. Sci 2018, 5, 1700540. [Google Scholar] [CrossRef] [PubMed]
  65. Yoshiba, T.; Saga, Y.; Urabe, M.; Uchibori, R.; Matsubara, S.; Fujiwara, H.; Mizukami, H. CRISPR/Cas9-mediated cervical cancer treatment targeting human papillomavirus E6. Oncol. Lett. 2018, 17, 2197–2206. [Google Scholar] [CrossRef]
  66. Li, C.; Guo, L.; Liu, G.; Guo, M.; Wei, H.; Yang, Q.; Wang, J.; Chen, H. Reprogrammed CRISPR-Cas13a Targeting the HPV16/18 E6 Gene Inhibits Proliferation and Induces Apoptosis in E6-Transformed Keratinocytes. Exp. Ther. Med. 2020, 19, 3856–3860. [Google Scholar] [CrossRef]
  67. Inturi, R.; Jemth, P. CRISPR/Cas9-Based Inactivation of Human Papillomavirus Oncogenes E6 or E7 Induces Senescence in Cer-Vical Cancer Cells. Virology 2021, 562, 92–102. [Google Scholar] [CrossRef]
  68. Noroozi, Z.; Shamsara, M.; Valipour, E.; Esfandyari, S.; Ehghaghi, A.; Monfaredan, A.; Azizi, Z.; Motevaseli, E.; Modarressi, M.H. Antiproliferative Effects of AAV-Delivered CRISPR/Cas9-Based Degradation of the HPV18-E6 Gene in HeLa Cells. Sci. Rep. 2022, 12, 2224. [Google Scholar] [CrossRef]
  69. Hu, Z.; Liu, W.; Liu, J.; Zhou, H.; Sun, C.; Tian, C.; Guo, X.; Zhu, C.; Shao, M.; Wang, S. The Anti-Tumor Efficacy of a Recom-Binant Oncolytic Herpes Simplex Virus Mediated CRISPR/Cas9 Delivery Targeting in HPV16-Positive Cervical Cancer. Antivir. Res. 2024, 232, 106035. [Google Scholar] [CrossRef]
  70. Zhang, A.; Zheng, X.; Chen, S.; Duan, G. In vitro study of HPV18-positive cervical cancer HeLa cells based on CRISPR/Cas13a system. Gene 2024, 921, 148527. [Google Scholar] [CrossRef]
  71. Shankar, S.; Prasad, D.; Sanawar, R.; Ani, V.; Pillai, M.R. TALEN Based HPV-E7 Editing Triggers Necrotic Cell Death in Cervical Cancer Cells. Sci. Rep 2017, 7, 5500. [Google Scholar] [CrossRef]
  72. Zhen, S.; Qiang, R.; Lu, J.; Tuo, X.; Yang, X.; Li, X. CRISPR/Cas9-HPV-Liposome Enhances Antitumor Immunity and Treatment of HPV Infection-Associated Cervical Cancer. J. Med. Virol. 2023, 95, 28144. [Google Scholar] [CrossRef]
  73. First Affiliated Hospital, Sun Yat-Sen University. A Safety and Efficacy Study of TALEN and CRISPR/Cas9 in the Treatment of HPV-Related Cervical Intraepithelial Neoplasia (NCT03057912). ClinicalTrials.gov.; 2017. Available online: https://clinicaltrials.gov/study/NCT03057912 (accessed on 8 October 2025).
  74. Sichuan University. PD-1 Knockout Engineered T Cells for Metastatic Non-Small Cell Lung Cancer (NCT02793856). ClinicalTrials.gov.; 2021. Available online: https://clinicaltrials.gov/study/NCT02793856 (accessed on 8 October 2025).
  75. Inovio Pharmaceuticals. Phase I of Human Papillomavirus (HPV) DNA Plasmid (VGX-3100) + Electroporation for CIN 2 or 3 (NCT00685412). ClinicalTrials.gov.; 2017. Available online: https://clinicaltrials.gov/study/NCT00685412 (accessed on 8 October 2025).
  76. Inovio Pharmaceuticals. A Study of VGX-3100 DNA Vaccine with Electroporation in Patients with Cervical Intraepithelial Neoplasia Grade 2/3 or 3 (HPV-003) (NCT01304524). ClinicalTrials.gov.; 2018. Available online: https://clinicaltrials.gov/study/NCT01304524 (accessed on 8 October 2025).
  77. Inovio Pharmaceuticals. Evaluation of VGX-3100 and Electroporation Alone or in Combination With Imiquimod for the Treatment of HPV-16 and/or HPV-18 Related Vulvar HSIL (Also Referred as: VIN 2 or VIN 3) (NCT03180684). ClinicalTrials.gov.; 2023. Available online: https://clinicaltrials.gov/study/NCT03180684 (accessed on 8 October 2025).
  78. Inovio Pharmaceuticals. VGX-3100 Delivered Intramuscularly (IM) Followed by Electroporation (EP) for the Treatment of HPV-16 and/or HPV-18 Related Anal or Anal/Peri-Anal, High Grade Squamous Intraepithelial Lesion (HSIL) in Individuals Seronegative for Human Immunodeficiency Virus (HIV)-1/2 (NCT03499795). ClinicalTrials.gov.; 2023. Available online: https://www.clinicaltrials.gov/study/NCT03499795 (accessed on 8 October 2025).
  79. VGX-3100 and Electroporation in Treating Patients with HIV-Positive High-Grade Anal Lesions (NCT03603808). ClinicalTrials.gov; 2025. Available online: https://clinicaltrials.gov/study/NCT03603808 (accessed on 8 October 2025).
  80. Kermanshahi, A.Z.; Ebrahimi, F.; Taherpoor, A.; Eslami, N.; Baghi, H.B. HPV-driven cancers: A looming threat and the potential of CRISPR/Cas9 for targeted therapy. Virol. J. 2025, 22, 156. [Google Scholar] [CrossRef]
  81. Wiik, J.; Kärrberg, C.; Nilsson, S.; Strander, B.; Jacobsson, B.; Sengpiel, V. Associations between cervical intraepithelial neoplasia during pregnancy, previous excisional treatment, cone-length and preterm delivery: A register-based study from western sweden. BMC Med. 2022, 20, 61. [Google Scholar] [CrossRef] [PubMed]
  82. Zhen, S.; Chen, H.; Lu, J.; Yang, X.; Tuo, X.; Chang, S.; Tian, Y.; Li, X. Intravaginal delivery for CRISPR-Cas9 technology: For example, the treatment of HPV infection. J. Med. Virol. 2023, 95, e28552. [Google Scholar] [CrossRef] [PubMed]
  83. Ding, W.; Cai, W.; Wang, H. P53 and pRB induction improves response to radiation therapy in HPV-positive laryngeal squamous cell carcinoma. Clinics 2024, 79, 100415. [Google Scholar] [CrossRef]
  84. Chung, H.C.; Ros, W.; Delord, J.P.; Perets, R.; Italiano, A.; Shapira-Frommer, R.; Manzuk, L.; Piha-Paul, S.A.; Xu, L.; Zeigenfuss, S.; et al. Efficacy and Safety of Pembrolizumab in Previously Treated Advanced Cervical Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2019, 37, 1470–1478. [Google Scholar] [CrossRef]
  85. Doran, S.L.; Stevanović, S.; Adhikary, S.; Gartner, J.J.; Jia, L.; Kwong, M.L.M.; Faquin, W.C.; Hewitt, S.M.; Sherry, R.M.; Yang, J.C.; et al. T-cell receptor gene therapy for human papillomavirus–associated epithelial cancers: A first-in-human, phase i/ii study. J. Clin. Oncol. 2019, 37, 2759–2768. [Google Scholar] [CrossRef]
  86. Liu, N.; Wang, X.; Wang, Z.; Kan, Y.; Fang, Y.; Gao, J.; Kong, X.; Wang, J. Nanomaterials-driven in situ vaccination: A novel frontier in tumor immunotherapy. J. Hematol. Oncol. 2025, 18, 45. [Google Scholar] [CrossRef]
  87. Zheng, S.; Wang, W.; Shen, L.; Yao, Y.; Xia, W.; Ni, C. Tumor battlefield within inflamed, excluded or desert immune phenotypes: The mechanisms and strategies. Exp. Hematol. Oncol. 2024, 13, 80. [Google Scholar] [CrossRef]
  88. García, E.; Ayoub, N.; Tewari, K.S. Recent breakthroughs in the management of locally advanced and recurrent/metastatic cervical cancer. J. Gynecol. Oncol. 2024, 35, e30. [Google Scholar] [CrossRef]
  89. Qi, L.; Luo, Q.; Zhang, Y.; Jia, F.; Zhao, Y.; Wang, F. Advances in toxicological research of the anticancer drug cisplatin. Chem. Res. Toxicol. 2019, 32, 1469–1486. [Google Scholar] [CrossRef] [PubMed]
  90. Viswanathan, A.N.; Lee, L.J.; Eswara, J.R.; Horowitz, N.S.; Konstantinopoulos, P.A.; Mirabeau-Beale, K.L.; Rose, B.S.; Von Keudell, A.G.; Wo, J.Y. Complications of pelvic radiation in patients treated for gynecologic malignancies. Cancer 2014, 120, 3870–3883. [Google Scholar] [CrossRef] [PubMed]
  91. Coventry, B.J. Therapeutic vaccination immunomodulation: Forming the basis of all cancer immunotherapy. Ther. Adv. Vaccines Immunother. 2019, 7, 2515135519862234. [Google Scholar] [CrossRef] [PubMed]
  92. Zolfi, E.; Khaleghi Mehr, F.; Emtiazi, N.; Moradi, Y. A review of the carcinogenic potential of human papillomavirus (HPV) in urological cancers. Virol. J. 2025, 22, 53. [Google Scholar] [CrossRef]
  93. Caley, A.; Evans, M.; Powell, N.; Paleri, V.; Tomkinson, A.; Urbano, T.G.; Jay, A.; Robinson, M.; Thavaraj, S. Multicentric human papillomavirus–associated head and neck squamous cell carcinoma. Head Neck 2014, 37, 202–208. [Google Scholar] [CrossRef]
  94. Khanna, L.; Prasad, S.R.; Yedururi, S.; Parameswaran, A.M.; Marcal, L.P.; Sandrasegaran, K.; Tirumani, S.H.; Menias, C.O.; Katabathina, V.S. Second malignancies after radiation therapy: Update on pathogenesis and cross-sectional imaging findings. Radiat. Oncol. 2021, 41, 876–894. [Google Scholar] [CrossRef]
  95. Zhou, P.; Liu, W.; Cheng, Y.; Qian, D. Nanoparticle-based applications for cervical cancer treatment in drug delivery, gene editing, and therapeutic cancer vaccines. WIREs Nanomed. Nanobiotechnol. 2021, 13, e1718. [Google Scholar] [CrossRef]
  96. Calderón, M.; Hedtrich, S. The delivery challenge of genome editing in human epithelia. Adv. Healthc. Mater. 2021, 10, 2100847. [Google Scholar] [CrossRef] [PubMed]
  97. Mutombo, A.B.; Simoens, C.; Tozin, R.; Bogers, J.; Van Geertruyden, J.P.; Jacquemyn, Y. Efficacy of commercially available biological agents for the topical treatment of cervical intraepithelial neoplasia: A systematic review. Syst. Rev. 2019, 8, 132. [Google Scholar] [CrossRef]
  98. Bahrami, K.; Lee, E.; Morse, B.; Lanier, O.L.; Peppas, N.A. Design of nanoparticle-based systems for the systemic delivery of chemotherapeutics: Alternative potential routes via sublingual and buccal administration for systemic drug delivery. Drug Deliv. Transl. Res. 2023, 14, 1173–1188. [Google Scholar] [CrossRef]
  99. Kotterman, M.A.; Chalberg, T.W.; Schaffer, D.V. Viral vectors for gene therapy: Translational and clinical outlook. Annu. Rev. Biomed. Eng. 2015, 17, 63–89. [Google Scholar] [CrossRef]
  100. Zhu, D.; Shen, H.; Tan, S.; Hu, Z.; Wang, L.; Yu, L.; Tian, X.; Ding, W.; Ren, C.; Gao, C.; et al. Nanoparticles Based on Poly (β-Amino Ester) and HPV16-Targeting CRISPR/shRNA as Potential Drugs for HPV16-Related Cervical Malignancy. Mol. Ther. 2018, 26, 2443–2455. [Google Scholar] [CrossRef]
  101. Gao, X.; Jin, Z.; Tan, X.; Zhang, C.; Zou, C.; Zhang, W.; Ding, J.; Das, B.C.; Severinov, K.; Hitzeroth, I.I.; et al. Hyperbranched poly(β-amino ester) based polyplex nanopaticles for delivery of CRISPR/Cas9 system and treatment of HPV infection associated cervical cancer. J. Control. Release 2020, 321, 654–668. [Google Scholar] [CrossRef]
  102. Xiong, J.; Tan, S.; Yu, L.; Shen, H.; Qu, S.; Zhang, C.; Ren, C.; Zhu, D.; Wang, H. E7-Targeted Nanotherapeutics for Key HPV Afflicted Cervical Lesions by Employing CRISPR/Cas9 and Poly (Beta-Amino Ester). Int. J. Nanomed. 2021, 16, 7609–7622. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, S.; Liu, Z.; Lai, L.; Li, Z. Efficient C-to-G Base Editing with Improved Target Compatibility Using Engineered Deaminase-nCas9 Fusions. Cris. J. 2022, 5, 389–396. [Google Scholar] [CrossRef]
  104. Wilbie, D.; Walther, J.; Mastrobattista, E. Delivery aspects of crispr/cas for in vivo genome editing. Acc. Chem. Res. 2019, 52, 1555–1564. [Google Scholar] [CrossRef] [PubMed]
  105. Mingozzi, F.; High, K.A. Overcoming the host immune response to adeno-associated virus gene delivery vectors: The race between clearance, tolerance, neutralization, and escape. Annu. Rev. Virol. 2017, 4, 511–534. [Google Scholar] [CrossRef]
  106. Cencic, R.; Miura, H.; Malina, A.; Robert, F.; Ethier, S.; Schmeing, T.M.; Dostie, J.; Pelletier, J. Protospacer adjacent motif (pam)-distal sequences engage crispr cas9 dna target cleavage. PLoS ONE 2014, 9, e109213. [Google Scholar] [CrossRef] [PubMed]
  107. Liu, Y.; Chao, Z.; Ding, W.; Fang, T.; Gu, X.; Xue, M.; Wang, W.; Han, R.; Sun, W. A Multiplex Rpa-Crispr/Cas12a-Based Poct Technique and Its Application in Human Papillomavirus (HPV) Typing Assay. Cell. Mol. Biol. Lett. 2024, 29, 34. [Google Scholar] [CrossRef]
  108. Mills, C.; Riching, A.; Keller, A.; Stombaugh, J.; Haupt, A.; Maksimova, E.; Dickerson, S.; Anderson, E.; Hemphill, K.; Ebmeier, C. A novel crispr interference effector enabling functional gene characterization with synthetic guide rnas. Cris. J. 2022, 5, 769–786. [Google Scholar] [CrossRef]
  109. Wu, Z.; Tang, Y.; Tang, M.; Wu, Z.; Xu, Y. The relationship between the eradication of helicobacter pylori and the occurrence of stomach cancer: An updated meta-analysis and systemic review. BMC Gastroenterol. 2025, 25, 278. [Google Scholar] [CrossRef]
  110. Aghamiri, S.; Talaei, S.; Roshanzamiri, S.; Zandsalimi, F.; Fazeli, E.; Aliyu, M.; Kheiry Avarvand, O.; Ebrahimi, Z.; Keshavarz-Fathi, M.; Ghanbarian, H. Delivery of genome editing tools: A promising strategy for HPV-related cervical malignancy therapy. Expert Opin. Drug Deliv. 2020, 17, 753–766. [Google Scholar] [CrossRef] [PubMed]
  111. Komor, A.C.; Badran, A.H.; Liu, D.R. CRISPR-Based Technologies for the Manipulation of Eukaryotic Genomes. Cell 2017, 168, 20–36. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA 2020 flow diagram of the study selection process. * Databases include PubMed, Scopus, and Web of Science. ** Records excluded: non-original, non-English, or unrelated to HPV/genome editing studies.
Figure 1. PRISMA 2020 flow diagram of the study selection process. * Databases include PubMed, Scopus, and Web of Science. ** Records excluded: non-original, non-English, or unrelated to HPV/genome editing studies.
Amh 70 00041 g001
Figure 2. CRISPR, TALENs & ZFNs in HPV Therapy. Schematic representation of genome editing strategies targeting high-risk HPV oncogenes E6 and E7. Viral genome integration into host DNA drives malignant transformation. Targeting HPV E6/E7 using CRISPR/Cas9, TALENs, or zinc-finger nucleases (ZFNs) induces double-strand breaks (DSBs) and knockout (KO) of viral sequences, leading to restoration of p53 and pRb tumor suppressor functions. This results in cell cycle arrest, growth inhibition, and induction of apoptosis in HPV-positive cancer cells.
Figure 2. CRISPR, TALENs & ZFNs in HPV Therapy. Schematic representation of genome editing strategies targeting high-risk HPV oncogenes E6 and E7. Viral genome integration into host DNA drives malignant transformation. Targeting HPV E6/E7 using CRISPR/Cas9, TALENs, or zinc-finger nucleases (ZFNs) induces double-strand breaks (DSBs) and knockout (KO) of viral sequences, leading to restoration of p53 and pRb tumor suppressor functions. This results in cell cycle arrest, growth inhibition, and induction of apoptosis in HPV-positive cancer cells.
Amh 70 00041 g002
Figure 3. Personalized genome editing strategies for HPV-associated cancers. (A) Genotype-specific guide RNA design improves precision by matching sequence variants of different HPV genotypes (e.g., HPV16 vs. HPV18). (B) Multiplexed CRISPR approaches enable simultaneous targeting of HPV oncogenes (E6/E7) and mutated host oncogenes identified by sequencing. (C) Precision targeting of patient-specific polymorphisms allows alternative guide RNA design to accommodate single-nucleotide polymorphisms (SNPs) within CRISPR target sites, enhancing editing accuracy for individualized therapy.
Figure 3. Personalized genome editing strategies for HPV-associated cancers. (A) Genotype-specific guide RNA design improves precision by matching sequence variants of different HPV genotypes (e.g., HPV16 vs. HPV18). (B) Multiplexed CRISPR approaches enable simultaneous targeting of HPV oncogenes (E6/E7) and mutated host oncogenes identified by sequencing. (C) Precision targeting of patient-specific polymorphisms allows alternative guide RNA design to accommodate single-nucleotide polymorphisms (SNPs) within CRISPR target sites, enhancing editing accuracy for individualized therapy.
Amh 70 00041 g003aAmh 70 00041 g003b
Table 1. Comparison of genome editing technologies for therapeutic targeting in HPV-related cancers.
Table 1. Comparison of genome editing technologies for therapeutic targeting in HPV-related cancers.
FeatureCRISPR/Cas9TALENsZFNs
MechanismRNA-guided DNA cleavage via Cas9 endonucleaseDNA-binding TALE repeats fused to FokI nucleaseZinc-finger domains fused to FokI nuclease
Target
Recognition
20-nt guide RNA binds complementary DNA adjacent to PAMTALE repeats recognize individual nucleotides, dimerize FokIZinc fingers recognize 3–4 bp each, FokI dimerization
Ease of DesignVery high → only guide RNA sequence needs to be changedModerate → modular but labor-intensive to assembleLow → requires protein engineering and optimization
SpecificityHigh with optimized guides, off-targets minimized using high-fidelity variantsVery high → one-to-one nucleotide recognition; no PAM requirementVariable → some designs highly specific, others very prone to off-targets
DeliveryChallenging due to Cas9 size (~4.2 kb), solved via smaller orthologs or mRNALarge plasmid size (>3 kb per TALEN), amenable to mRNA or protein deliverySmallest, can fit both ZFNs in one AAV vector
MultiplexingYes → multiple sgRNAs can be used simultaneouslyDifficult → requires multiple TALEN pairsVery difficult → requires multiple protein constructs
Off-target RiskModerate → reduced with proper guide design and variantsLow → mismatch discrimination is highHigh → may cut hundreds of off-target sites depending on design
This table summarizes key features of the three major genome editing platforms—CRISPR/Cas9, TALENs, and ZFNs—focusing on their mechanisms, design simplicity, specificity, delivery challenges, and relevance to HPV research. While CRISPR/Cas9 offers the greatest ease of use and scalability, TALENs provide high specificity without PAM constraints, and ZFNs, though pioneering, are limited by complex design and higher off-target potential. These comparisons help guide the selection of appropriate editing tools for clinical translation in HPV-driven malignancies.
Table 2. Selected In Vitro Preclinical Studies Using Genome Editing in HPV Models.
Table 2. Selected In Vitro Preclinical Studies Using Genome Editing in HPV Models.
Gene Editing ToolLesion TypeCell LineTarget GeneOutcomes/FindingsReference
CRISPR/Cas9Cervical cancerSiHa, CaSkiHPV16 E7
  • Apoptosis: 40–56% in SiHa, 35–48% in CaSki
  • gRNA-4 showed strongest reduction in viability
Hu et al., 2014 [14]
CRISPR/Cas9Cervical cancerHela, SiHaHPV18 E6 & E7
  • ↓E6/E7 mRNA & protein (Hela)
  • ↑p53/p21 expression
  • ↑Apoptosis & ↓proliferation
Ling et al., 2020 [22]
CRISPR/Cas9Genital wartsHPV6/11 E7-transformed keratinocytesHPV6/11 E7
  • Induced apoptosis in E7-transformed keratinocytes
Liu et al., 2016 [63]
CRISPR/Cas9Cervical cancerSiHa, C33AHPV16 E6 & E7
  • Reduced viability in SiHa, minor effect in C33A
  • Combined with CDDP enhanced growth inhibition
Zhen et al., 2016 [41]
CRISPR/Cas9 (micelles)Cervical cancerHeLaHPV18 E7
  • ↓Proteasome activity (25–32%)
  • ↓Cell growth (68–73%)
  • ↑Rb expression
Lao et al., 2018 [64]
CRISPR/Cas9 (AAV)Cervical cancerHeLa, HCS-2, SKG-I, HEK293HPV18 E6
  • ↑Apoptosis (~46 FITC+ cells)
  • Dose-dependent growth suppression
Yoshiba et al., 2018 [65]
CRISPR/Cas9, FokI-dCas9Cervical cancerCaSki, HeLaHPV16/18 E6 & E7
  • ↑p53, p21, Rb
  • WT Cas9 > FokI-dCas9 in protein restoration
Jubair et al., 2019 [52]
CRISPR/Cas9 (HCAdV)Cervical cancerSiHa, CaSki, HeLaHPV16/18 E6
  • ↓Viability and ↑apoptosis in HPV+ cells
  • No effect in HPV-negative cells
Ehrke-Schulz et al., 2020 [42]
CRISPR/Cas13aCervical cancerSiHa, HeLa, C33AHPV16/18 E6 & E7
  • Knockdown of E6/E7 mRNA
  • ↑p53 and Rb in HPV+ cells only
Chen et al., 2020 [54]
CRISPR/Cas13aGenital wartsHPV16/18 E6-transformed keratinocytesHPV16/18 E6
  • ↓Proliferation (−14%)
  • ↑Caspase-3 (80%)
  • ↑Hoechst positivity
Li et al., 2020 [66]
CRISPR/Cas9Cervical cancerHeLaHPV18 E6 & E7
  • Triggered senescence
  • ↑β-galactosidase, ↓lamin B1
  • ↑p53/p21 and pRb/p21
Inturi et al., 2021 [67]
CRISPR/Cas9 (inducible)Oral SCCUDSCC2 (SCC2)HPV16 E7
  • E7 deletion regressed tumors
  • Doxycycline-induced expression
Ferreira et al., 2022 [56]
CRISPR/Cas9, TALEN, ZFNCervical cancerSiHa, S12, HeLa, C33AHPV16 E7
  • CRISPR most effective
  • ↓E7, ↑Rb, ↓E2F1/CDK2
  • Minimal effect in HeLa/C33A
Gao et al., 2022 [43]
CRISPR/Cas9 (AAV)Cervical cancerHeLaHPV18 E6
  • ↑p53 expression
  • ↑Apoptosis up to 53% with dual sgRNAs
Noroozi et al., 2022 [68]
CRISPR/Cas9Cervical cancerC3, TC1, HeLaHPV16 E5, E6, E7
  • ↓Oncoproteins in C3
  • ↑p21 (E5), ↑p53 (E6), ↑Rb (E7)
  • No effect in HeLa (HPV18+)
Khairkhah et al., 2023 [53]
CRISPR/Cas9 (SONC103)Cervical cancerCaSkiHPV16 E6, E7, E2, E5
  • ↓E6/E7, ↑p53 and pRb
  • ↑Apoptosis
Hu et al., 2024 [69]
CRISPR/Cas13aCervical cancerHeLaHPV18 E6
  • ↓HPV18 E6 mRNA
  • ↑p53, ↑Apoptosis
  • Enhanced cisplatin effect
Zhang et al., 2024 [70]
TALENCervical cancerHeLa, SiHa, C33A, HEK293HPV16/18 E6 & E7
  • Specific apoptosis in HPV-matched lines
  • No toxicity in C33A, HEK293
Hu et al., 2015 [55]
TALENCervical cancerSiHaHPV16 E7
  • Induced necrotic morphology, not apoptosis
Shankar et al., 2017 [71]
ZFNCervical cancerSiHa, HeLaHPV16/18 E7
  • Disrupted E7 oncogene
  • Restored RB1 pathway
Ding et al., 2014 [36]
Abbreviations: ↑, increased; ↓, decreased.
Table 3. Selected In Vivo Preclinical Studies Using Genome Editing in HPV Models.
Table 3. Selected In Vivo Preclinical Studies Using Genome Editing in HPV Models.
Gene Editing ToolLesion TypeCell LineTarget GeneOutcomes/FindingsReference
CRISPR/Cas9Cervical cancerNude mice (HeLa xenograft)HPV18 E6 & E7
  • Tumor formation delayed in Cas9-treated mice
Ling et al., 2020 [22]
CRISPR/Cas9 + CDDPCervical cancerBALB/c nude mice (hydrodynamic tail vein)HPV16 E6 & E7
  • ↓Tumor volume, ↑p53/Bax, ↓Bcl-2
  • ↓Lung metastasis
Zhen et al., 2016 [41]
CRISPR/Cas9 (micelles)Cervical cancerNude mice (HeLa xenograft)HPV18 E7
  • ↓Tumor volume by ~60%
  • ↑Rb restoration, ↑necrosis
  • No systemic toxicity
Lao et al., 2018 [64]
CRISPR/Cas9 (AAV)Anal cancerImmunodeficient mice (ANA001 PDX)HPV16 E6 & E7
  • >2-fold tumor volume reduction
Hsu et al., 2018 [62]
CRISPR/Cas9 (AAV)Cervical cancerBALB/c nude mice (SKG-I xenograft + AAV-sgE6)HPV18 E6
  • Significant tumor suppression vs. control
Yoshiba et al., 2018 [65]
CRISPR/Cas9Cervical cancerRag1 mice (CaSki, HeLa xenograft, lipoplex Cas9)HPV16/18 E7
  • ↑Apoptosis, ↑necrosis
  • ↑Survival with 3 doses, full remission with 7 doses
Jubair et al., 2019 [52]
CRISPR/Cas9 + anti-PD1 immunotherapyCervical cancerhu-PBL-SCID (SiHa-Luc xenograft)HPV16 E6 & E7 + PD1
  • Modest effect with CRISPR alone
  • Significant survival gain with combo
Zhen et al., 2020 [48]
CRISPR/Cas13aCervical cancerBALB/c nude (SiHa/HeLa CRISPR knockdown)HPV16 E6
  • ↓Tumor weight/volume in CRISPR group
Chen et al., 2020 [54]
Inducible CRISPR/Cas9Oral SCCNude mice (SCC2 xenograft)HPV16 E7
  • Tumor regression with E7 deletion
  • No toxicity
Ferreira et al., 2022 [56]
CRISPR/Cas9Cervical cancerBALB/c nude (S12 xenograft)HPV16 E7
  • ↓Tumor size, ↓E7, CD31, PCNA
  • ↑Caspase-3
Gao et al., 2022 [43]
CRISPR/Cas9Cervical cancerK14-HPV16 transgenic miceHPV16 E7
  • ↓E7 and P16 expression
  • Reversal of CIN
Gao et al., 2022 [43]
CRISPR/Cas9 + liposomesCervical cancerhu-PBL-SCID (SiHa xenograft)HPV16 E6 & E7
  • ↑Autophagy, ↑DAMPs, ↓immunosuppressive TME, ↑ICB effect
Zhen et al., 2023 [72]
CRISPR/Cas9 + cisplatinCervical cancerC57BL/6 (C3 xenograft)HPV16 E5, E6, E7
  • ↑Cleaved caspase-3 (45.8%)
  • ↓Mitotic index
  • LL-37 improved delivery
Khairkhah et al., 2023 [53]
CRISPR/Cas9 (SONC103 virus)Cervical cancerBALB/c (CaSki xenograft)HPV16 E6, E7, E2, E5
  • ↓E6/E7, ↑p53, ↑pRb
  • ↑Apoptosis in TUNEL
Hu et al., 2024 [69]
TALENCervical cancerBALB/c nude + K14-HPV16 topical T512HPV16/18 E6 & E7
  • ↓Tumor size, limited vaginal/cervical epithelium effect
Hu et al., 2015 [55]
ZFNCervical cancerBALB/c nude (SiHa/HeLa xenografts)HPV16/18 E7
  • ↓Tumor growth and size
Ding et al., 2014 [36]
Abbreviations: ↑, increased; ↓, decreased.
Table 4. Ongoing Clinical Trial of Genome Editing Therapy in HPV-Related Disease. This first-in-human trial is testing local application of gene editing plasmids to cervical lesions. Its outcomes will shed light on safety (e.g., local inflammation, off-target mutations in patients) and provide initial signals of efficacy such as HPV clearance or lesion regression. Success here could pave the way for trials in higher-grade lesions or even early cancers.
Table 4. Ongoing Clinical Trial of Genome Editing Therapy in HPV-Related Disease. This first-in-human trial is testing local application of gene editing plasmids to cervical lesions. Its outcomes will shed light on safety (e.g., local inflammation, off-target mutations in patients) and provide initial signals of efficacy such as HPV clearance or lesion regression. Success here could pave the way for trials in higher-grade lesions or even early cancers.
TrialTarget/StrategyIndicationApproach
NCT03057912 CRISPR/Cas9 or TALENs targeting HPV16/18 E6/E7CIN I with persistent HPV16/18 infectionIntravaginal plasmid delivery, open-label Phase I trial
NCT02793856 PD-1 knockout via CRISPR in autologous T cellsHPV+ metastatic non-small cell lung cancerT cell gene editing and reinfusion
NCT03578406 TCR-engineered T cells recognizing HPV16 E6Metastatic cervical cancerTCR-transduced T cells (not gene-edited yet)
NCT00685412
NCT01304524
NCT03185013
(REVEAL1)
NCT03721978
(REVEAL2)
VGX-3100 therapeutic vaccine targeting HPV16/18 E6/E7 DNACervical intraepithelial neoplasia (CIN) 2/3DNA vaccine + Electroporation
Phase I, II and III trials
NCT03603808VGX-3100 therapeutic vaccine targeting HPV16/18 E6/E7 DNAAnal high-grade squamous intraepithelial neoplasia (HSIL) or AIN 2/3 in HIV+ individualsDNA vaccine + Electroporation
Phase II trial
NCT03499795VGX-3100 therapeutic vaccine targeting HPV16/18 E6/E7 DNAAIN2, AIN3, PAIN2, PAIN3 in HIV– individualsDNA vaccine + Electroporation Phase II trial
NCT03180684VGX-3100 therapeutic vaccine targeting HPV16/18 E6/E7 DNAVulvar high-grade squamous intraepithelial lesion (HSIL) or VIN 2/3DNA vaccine + Electroporation
Phase II trial
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

Cakir, M.O.; Selek, M.; Yilmaz, B.; Ozdogan, M.; Ashrafi, G.H. Genome Editing Against HPV-Driven Cancers: From Bench to Clinic. Acta Microbiol. Hell. 2025, 70, 41. https://doi.org/10.3390/amh70040041

AMA Style

Cakir MO, Selek M, Yilmaz B, Ozdogan M, Ashrafi GH. Genome Editing Against HPV-Driven Cancers: From Bench to Clinic. Acta Microbiologica Hellenica. 2025; 70(4):41. https://doi.org/10.3390/amh70040041

Chicago/Turabian Style

Cakir, Muharrem Okan, Melis Selek, Betul Yilmaz, Mustafa Ozdogan, and Gholam Hossein Ashrafi. 2025. "Genome Editing Against HPV-Driven Cancers: From Bench to Clinic" Acta Microbiologica Hellenica 70, no. 4: 41. https://doi.org/10.3390/amh70040041

APA Style

Cakir, M. O., Selek, M., Yilmaz, B., Ozdogan, M., & Ashrafi, G. H. (2025). Genome Editing Against HPV-Driven Cancers: From Bench to Clinic. Acta Microbiologica Hellenica, 70(4), 41. https://doi.org/10.3390/amh70040041

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop