Recent Advances in Treatment Options for Locally Advanced Basal Cell Carcinoma: A Review of TVEC and Radiation Therapy

Abstract

Basal cell carcinoma (BCC) is the most common form of skin cancer worldwide, with the majority of cases being amenable to simple therapeutic interventions. However, a subset of patients presents with locally advanced BCC (laBCC), which poses significant therapeutic challenges due to tumor size, location, and potential for functional and cosmetic morbidity. This review examines recent advances in the management of laBCC, with a particular focus on two promising approaches: Talimogene Laherparepvec (TVEC) as a neoadjuvant therapy and definitive radiation therapy. Recent data from phase II clinical trials demonstrate that TVEC, a genetically modified oncolytic herpes simplex virus, can significantly reduce tumor size and even achieve complete pathological regression in some patients with laBCC. Meanwhile, multi-institutional data on definitive radiation therapy show high rates of locoregional control in patients with unresectable or functionally challenging laBCC. This paper reviews the evidence for both approaches, explores their respective advantages and limitations, and considers their place in the evolving treatment landscape for laBCC, which increasingly emphasizes multidisciplinary care. The emerging data suggest that these modalities, alone or in combination with other treatments, may provide effective alternatives to extensive surgery for patients with laBCC, potentially reducing functional and cosmetic morbidity while maintaining oncologic efficacy.

1. Introduction

Basal cell carcinoma (BCC) is the most prevalent form of skin cancer, accounting for approximately 80% of all non-melanoma skin cancers (Krishna et al., 2022). While the majority of BCCs are small, indolent, and readily amenable to surgical excision or local destructive techniques, a subset presents as locally advanced disease, defined by size (≥4 cm), depth of invasion, anatomic location, or recurrence despite prior therapy (Kim et al., 2024). These locally advanced basal cell carcinomas (laBCCs) present unique therapeutic challenges, especially when located in cosmetically and functionally sensitive areas such as the face, where extensive surgery may result in significant disfigurement or functional impairment (Koyfman et al., 2025).

Historically, wide local excision has been the standard of care for laBCC, often necessitating complex reconstruction with skin grafts or flaps (Vidimos et al., 2025). However, this approach may be associated with considerable morbidity, particularly for elderly patients or those with tumors in aesthetically sensitive areas. The limitations of surgery have prompted investigations into alternative or complementary therapeutic modalities, including systemic therapies targeting the Hedgehog signaling pathway, radiation therapy, and more recently, immunotherapeutic approaches (Höller et al., 2025).

In recent years, two approaches have emerged as particularly promising for the management of laBCC: (1) Talimogene Laherparepvec (TVEC), a genetically modified oncolytic herpes simplex virus, used as neoadjuvant therapy before planned surgical excision; and (2) definitive radiation therapy for unresectable tumors or cases where surgery would result in excessive morbidity. Both approaches aim to achieve local control while minimizing functional and cosmetic sequelae.

This review examines the current evidence for TVEC and definitive radiation therapy in the management of laBCC, with a focus on recent clinical trials and multi-institutional studies. Additionally, it explores the role of these modalities within the broader context of multidisciplinary care for patients with laBCC and identifies areas for future research and clinical development.

2. Pathophysiology and Clinical Features of Locally Advanced Basal Cell Carcinoma

2.1 Pathophysiology

Basal cell carcinoma arises from the basal layer of the epidermis and its appendages. The majority of BCCs are driven by aberrant activation of the Hedgehog signaling pathway, frequently through loss-of-function mutations in the tumor suppressor gene PTCH1 or gain-of-function mutations in the SMO gene (Atwood et al., 2021). This pathway plays a critical role in embryonic development and is normally suppressed in adult tissues. Its inappropriate activation leads to uncontrolled proliferation of basal cells and tumor formation.

Chronic ultraviolet (UV) radiation exposure is the primary environmental risk factor for BCC development, with approximately 80% of cases occurring in chronically sun-exposed areas such as the face and neck (Medical University of Vienna, 2025). The cumulative DNA damage induced by UV radiation contributes to the genetic alterations driving BCC pathogenesis.

Locally advanced BCCs typically result from neglected primary tumors or recurrent disease after prior treatment. They may exhibit aggressive behavior characterized by local tissue destruction, invasion of deeper structures including cartilage and bone, and perineural spread (Koyfman et al., 2025). Unlike metastatic BCC, which is rare (occurring in less than 0.1% of cases), laBCC presents challenges primarily related to local tumor control and functional preservation rather than systemic disease management.

2.2 Clinical Features and Diagnosis

Locally advanced BCCs typically present as large (≥4 cm), ulcerated lesions with rolled borders. Depending on the location, they may invade adjacent structures such as the orbit, nasal cavity, or ear canal, potentially causing functional impairment (Vidimos et al., 2025). Perineural invasion, which occurs in approximately 5-10% of cases, may manifest as pain, paresthesia, or motor deficits along the distribution of the affected nerve (Koyfman et al., 2025).

Diagnosis is established through skin biopsy, typically a punch or incisional biopsy to sample an adequate depth of tissue. Histopathological examination reveals characteristic features of BCC, including basaloid cells with peripheral palisading, retraction artifact, and a mucinous stroma. Various histological subtypes exist, with infiltrative, morpheaform, and basosquamous subtypes generally considered more aggressive (Kim et al., 2023).

For laBCC, comprehensive imaging is essential to delineate the extent of disease and guide treatment planning. Magnetic resonance imaging (MRI) is particularly valuable for assessing soft tissue involvement and perineural spread, while computed tomography (CT) may better visualize bone invasion. High-frequency ultrasonography can provide detailed information about tumor thickness and depth, aiding surgical planning (Koyfman et al., 2025).

3. Conventional Management Approaches for Locally Advanced BCC

Before discussing the emerging roles of TVEC and definitive radiation therapy, it is important to review the established approaches to laBCC management, which form the foundation upon which newer strategies are built.

3.1 Surgical Management

Surgery remains the gold standard for most BCCs, with several techniques employed depending on tumor characteristics and location:

  1. Wide Local Excision: The traditional approach, involving removal of the tumor with a margin of normal-appearing skin, typically 4-10 mm for laBCC (Vidimos et al., 2025).
  2. Mohs Micrographic Surgery: A specialized technique involving sequential removal and immediate microscopic examination of tissue until clear margins are achieved. This tissue-sparing approach is particularly valuable for facial lesions and recurrent tumors (Koyfman et al., 2025).
  3. Reconstruction: For laBCC, extensive surgical defects often necessitate complex reconstruction using local flaps, skin grafts, or free tissue transfer (Vidimos et al., 2025).

While surgery offers high cure rates, laBCC often presents in locations where complete excision with adequate margins may result in significant functional or cosmetic morbidity. Additionally, some patients may be poor surgical candidates due to advanced age, comorbidities, or tumor characteristics (Koyfman et al., 2025).

3.2 Hedgehog Pathway Inhibitors

The discovery of the central role of Hedgehog pathway dysregulation in BCC pathogenesis led to the development of targeted therapies, specifically Hedgehog pathway inhibitors (HPIs):

  1. Vismodegib: The first FDA-approved HPI for laBCC, vismodegib binds to and inhibits SMO, a key mediator of Hedgehog signaling. Clinical trials have demonstrated response rates of 30-60% in laBCC, with complete responses in approximately one-third of cases (Koyfman et al., 2025).
  2. Sonidegib: A second-generation HPI with a similar mechanism of action and efficacy profile to vismodegib.

While HPIs have revolutionized the management of inoperable laBCC, their use is limited by significant side effects, including muscle spasms, taste disturbance, alopecia, and weight loss, which lead to treatment discontinuation in up to 30% of patients. Additionally, tumor resistance typically develops within 6-12 months of therapy initiation (Koyfman et al., 2025).

3.3 Conventional Radiation Therapy

Radiation therapy has historically been used as a definitive treatment for BCC in patients unsuitable for surgery or as adjuvant therapy after incomplete resection. Conventional approaches include:

  1. External Beam Radiotherapy: Typically delivered in fractionated courses over 3-6 weeks, with total doses of 45-70 Gy depending on tumor characteristics (Koyfman et al., 2025).
  2. Brachytherapy: Involving the placement of radioactive sources directly into or adjacent to the tumor, allowing for high local doses while sparing surrounding tissues.

Traditional approaches to radiation therapy were limited by concerns about cosmetic outcomes, particularly late radiation toxicity, and technical challenges in treating complex, three-dimensional targets (Koyfman et al., 2025).

4. Talimogene Laherparepvec (TVEC) for Locally Advanced BCC

4.1 Mechanism of Action

Talimogene Laherparepvec (TVEC) represents a novel approach to cancer therapy as an oncolytic virus immunotherapy. TVEC is a genetically modified herpes simplex virus type 1 (HSV-1) designed to selectively replicate within tumor cells while sparing normal tissues (Ressler et al., 2025). Two key genetic modifications underlie its therapeutic activity:

  1. Deletion of ICP34.5 genes: Confers tumor selectivity by preventing viral replication in normal cells while permitting propagation in cancer cells with defective anti-viral responses.
  2. Insertion of GM-CSF gene: Enables the virus to express granulocyte-macrophage colony-stimulating factor (GM-CSF), a potent activator of antigen-presenting cells that enhances anti-tumor immune responses.

When injected directly into tumor tissue (intralesional administration), TVEC selectively infects and replicates within tumor cells, leading to:

  • Direct oncolysis (lysis of tumor cells)
  • Release of tumor-associated antigens
  • Local GM-CSF production
  • Recruitment and activation of dendritic cells and other immune effectors
  • Initiation of systemic anti-tumor immunity that may affect uninjected tumors (abscopal effect)

This dual mechanism—direct oncolysis plus immune activation—distinguishes TVEC from conventional therapies and provides a rationale for its investigation in laBCC (Höller et al., 2025).

4.2 Clinical Evidence for TVEC in laBCC

Until recently, TVEC was approved only for the treatment of unresectable melanoma, based on the phase III OPTiM trial, which demonstrated improved durable response rates compared to GM-CSF alone. However, new evidence suggests efficacy in laBCC as well.

The most significant contribution to this evidence comes from the NeoBCC trial, a phase II study conducted by researchers from the Medical University of Vienna and University Hospital Vienna. This trial investigated TVEC as neoadjuvant therapy for laBCC with the primary goal of reducing tumor size prior to planned surgical excision (Ressler et al., 2025; Medical University of Vienna, 2025).

Study Design and Population

The NeoBCC trial enrolled 18 patients with laBCC who, due to tumor size and location, would have required complex surgical closure with a flap or skin graft. Key inclusion criteria included:

  • Histologically confirmed BCC
  • Tumor size and location necessitating complex reconstruction
  • No prior systemic therapy for BCC

Patients received six intralesional injections of TVEC over a 13-week period prior to planned surgical resection (Ressler et al., 2025).

Efficacy Outcomes

The results of the NeoBCC trial demonstrated remarkable efficacy:

  1. Tumor Size Reduction: All treated tumors (100%) showed reduction in size, with no cases of progression during therapy.
  2. Surgical Outcomes: In 50% of patients, tumor shrinkage was sufficient to permit direct wound closure without the need for complex reconstruction, significantly reducing surgical morbidity.
  3. Pathological Complete Response: In approximately one-third (33%) of patients, subsequent histological examination of the surgical specimen revealed no viable tumor cells, indicating complete pathological response to TVEC therapy alone (Höller et al., 2025; Medical University of Vienna, 2025).

These findings are particularly significant given the challenges of treating laBCC in functionally and cosmetically sensitive areas, where extensive surgery may result in disfigurement or functional impairment.

Safety and Tolerability

The NeoBCC trial reported that TVEC therapy was generally well-tolerated. The most common adverse events were consistent with the known safety profile of TVEC in melanoma, including:

  • Flu-like symptoms (fever, chills, fatigue)
  • Local injection site reactions
  • Transient lymphadenopathy

No treatment-related serious adverse events were reported, and all patients completed the planned course of therapy (Ressler et al., 2025).

4.3 Immunological Effects

Beyond the clinical outcomes, the NeoBCC trial provided insights into the immunological effects of TVEC therapy in laBCC. Comprehensive analyses conducted in collaboration with the St. Anna Children's Hospital demonstrated:

  1. Enhanced Immune Infiltration: Increased presence of T cells and other immune effectors within the tumor microenvironment following TVEC therapy.
  2. Activation of Anti-tumor Immunity: Evidence of immune activation and improved immune defense in tumor tissue during TVEC therapy (Medical University of Vienna, 2025).

These findings support the proposed mechanism of action of TVEC as both a direct oncolytic agent and an immunotherapy, capable of converting immunologically "cold" tumors to "hot" tumors more susceptible to immune-mediated clearance.

5. Definitive Radiation Therapy for Locally Advanced BCC

5.1 Evolution of Radiation Techniques

While radiation therapy has been used for decades in the management of BCC, recent advances in technology and delivery techniques have substantially improved its efficacy and tolerability for laBCC:

  1. Intensity-Modulated Radiation Therapy (IMRT): Allows for precise shaping of radiation dose to conform to complex target volumes while minimizing exposure to adjacent normal tissues.
  2. Volumetric Arc Therapy (VMAT): Enables rapid and accurate delivery of radiation from multiple angles, improving dose homogeneity and reducing treatment time.
  3. Image-Guided Radiation Therapy (IGRT): Incorporates daily imaging to ensure accurate targeting throughout the treatment course, particularly valuable for facial lesions where millimeter precision is essential.

These advances have dramatically reduced side effects and expanded the role of radiation therapy in managing complex laBCC cases (Koyfman et al., 2025).

5.2 Clinical Evidence for Definitive Radiation Therapy

A significant contribution to the evidence supporting definitive radiation therapy for laBCC comes from a recent multi-institutional study conducted by Cleveland Clinic Foundation, the University of Pennsylvania Health System, and Brigham and Women's Hospital (Koyfman et al., 2025).

Study Design and Population

This retrospective study identified 680 patients with laBCC treated between 2005 and 2021, of whom 140 received definitive radiation therapy. For this study, "locally advanced" was defined as tumors that were:

  • ≥4 cm in size
  • Required extensive resection
  • Deemed unresectable
  • Would have required neoadjuvant therapy prior to resection

The study population included 101 patients treated with radiation at initial diagnosis and 39 with recurrent disease after prior therapy. Most patients (70.4%) were treated with electron-based radiation plans, with dose fractionation regimens ranging from hypofractionated (30-35 Gy in five fractions over two weeks) to conventional fractionation (60-70 Gy in 30-35 fractions over 6-7 weeks) (Koyfman et al., 2025).

Efficacy Outcomes

The results demonstrated high efficacy of definitive radiation therapy:

  1. Locoregional Control: Five-year Kaplan-Meier estimates of locoregional control exceeded 75% (78% specifically), indicating durable tumor control in a majority of patients.
  2. Recurrence Patterns: At a median follow-up of 22.9 months, 26 patients (18.6%) developed recurrence, with 22 (84.6%) being locoregional failures and only 4 (15.4%) representing distant metastases. The median time to recurrence was 25.8 months.
  3. Disease-Specific Survival: Five-year BCC-specific survival was 85%, with similar outcomes in patients treated with upfront definitive radiation (82.7%) compared to those receiving radiation for recurrent disease (86.7%).
  4. Prognostic Factors: Subset analysis identified a high-risk group characterized by one or more adverse features (size ≥4 cm, bone invasion, perineural invasion, immunocompromised status, or recurrent disease), which had a significantly lower five-year freedom from locoregional failure rate (68.5% versus 92.4% in patients without these features) (Koyfman et al., 2025).

These findings represent one of the largest contemporary studies demonstrating the efficacy of definitive radiation therapy for laBCC and provide valuable prognostic information for treatment selection and patient counseling.

5.3 Advantages and Limitations

Definitive radiation therapy offers several advantages for patients with laBCC:

  1. Non-invasive Treatment: Avoids the morbidity associated with extensive surgery, particularly important for elderly patients or those with comorbidities.
  2. Tissue Preservation: Maintains normal tissue architecture and function, potentially resulting in superior cosmetic and functional outcomes compared to extensive surgery.
  3. Applicability to Complex Cases: Effective for tumors in difficult anatomic locations or with perineural spread, which may be challenging to address surgically.

However, limitations should also be acknowledged:

  1. Treatment Duration: Conventional fractionation regimens require daily treatments over 6-7 weeks, which may be logistically challenging for some patients.
  2. Late Radiation Effects: Risk of long-term sequelae, including fibrosis, telangiectasia, and, rarely, secondary malignancies, though modern techniques have significantly reduced these risks.
  3. Contraindications: Not suitable for patients with certain genetic conditions predisposing to skin cancer (e.g., Gorlin syndrome) due to increased risk of radiation-induced malignancies (Koyfman et al., 2025).

6. Multidisciplinary Approach to Locally Advanced BCC

6.1 Importance of Multidisciplinary Care

The management of laBCC increasingly emphasizes a multidisciplinary approach, bringing together expertise from multiple specialties to optimize treatment strategies. This approach is particularly valuable given the expanding therapeutic options and the complex decision-making required for patients with challenging tumors (Vidimos et al., 2025).

Key specialists involved typically include:

  • Dermatologists
  • Mohs surgeons
  • Surgical/plastic reconstructive surgeons
  • Radiation oncologists
  • Medical oncologists
  • Pathologists
  • Radiologists

Institutional models, such as that employed at Cleveland Clinic, demonstrate the value of multidisciplinary tumor boards specifically focused on non-melanoma skin cancer. These forums facilitate:

  1. Comprehensive Case Assessment: Considering all aspects of the tumor, patient characteristics, and treatment options.
  2. Sequential Treatment Planning: Determining optimal sequencing of multimodality therapy (e.g., neoadjuvant systemic therapy followed by surgery or radiation).
  3. Ongoing Reassessment: Adapting treatment plans based on tumor response and patient tolerance (Koyfman et al., 2025).

6.2 Treatment Selection and Sequencing

The expanding therapeutic armamentarium for laBCC necessitates careful consideration of treatment selection and sequencing, guided by:

  1. Tumor Characteristics: Size, location, histological subtype, presence of adverse features (bone invasion, perineural spread).
  2. Patient Factors: Age, comorbidities, functional status, patient preferences regarding treatment goals and acceptable morbidity.
  3. Previous Treatments: History of prior interventions and their outcomes.

For many patients with laBCC, combination approaches may offer the best chance for optimal oncologic and functional outcomes. Emerging paradigms include:

  1. TVEC + Surgery: As demonstrated in the NeoBCC trial, neoadjuvant TVEC can reduce tumor size sufficiently to permit less extensive surgery with improved functional and cosmetic outcomes.
  2. Systemic Therapy + Surgery/Radiation: Hedgehog pathway inhibitors may be used to downstage tumors prior to definitive local therapy, potentially improving resectability or radiation targeting.
  3. Surgery + Adjuvant Radiation: For high-risk features (positive margins, perineural invasion) identified on surgical pathology (Koyfman et al., 2025).
  4. Combination Systemic Approaches: For example, combining hedgehog inhibitors with immune checkpoint inhibitors for particularly challenging cases.

6.3 Overcoming Barriers to Care

The multidisciplinary approach also addresses important barriers to optimal care for patients with laBCC:

  1. Access to Specialized Expertise: Through telemedicine consultations and virtual tumor boards, expertise can be extended beyond tertiary centers to community settings.
  2. Financial Considerations: Navigation services can help address insurance and cost concerns, particularly for novel therapies like TVEC.
  3. Geographical Barriers: Development of regional referral networks can facilitate appropriate referrals while minimizing travel burden for patients (Vidimos et al., 2025).

7. Future Directions

7.1 Ongoing Clinical Trials

The management of laBCC continues to evolve, with several important ongoing investigations:

  1. Expanded TVEC Studies: Building on the promising results of the NeoBCC trial, larger studies are planned to confirm the efficacy of neoadjuvant TVEC and identify optimal candidates for this approach.
  2. Novel Radiation Approaches: Investigation of ultra-hypofractionated regimens (1-5 fractions) using stereotactic techniques to reduce treatment duration while maintaining efficacy.
  3. Immune Checkpoint Inhibitors: Exploration of anti-PD-1/PD-L1 therapies, alone or in combination with other modalities, for laBCC, particularly for patients with immunogenic tumor subtypes.
  4. Biomarker Development: Identification of molecular and immunological predictors of response to specific therapies, enabling more personalized treatment selection.

7.2 Molecular and Immune Monitoring

Advances in tumor molecular profiling and immune monitoring may provide valuable tools for treatment selection and response assessment:

  1. Genetic Profiling: Beyond PTCH1 and SMO mutations, comprehensive genomic analysis may identify additional therapeutic targets and resistance mechanisms.
  2. Immune Landscape Assessment: Characterization of the tumor immune microenvironment may help predict response to immunotherapeutic approaches, including TVEC.
  3. Liquid Biopsies: Detection of circulating tumor DNA may provide non-invasive means for monitoring treatment response and early detection of recurrence.

7.3 Novel Therapeutic Approaches

Several emerging therapies show promise for future management of laBCC:

  1. Next-Generation Hedgehog Inhibitors: Designed to overcome resistance to first-generation agents or with improved side effect profiles.
  2. Novel Oncolytic Viruses: Building on the success of TVEC, additional engineered viral platforms are under investigation.
  3. Targeted Radionuclide Therapy: Coupling of radionuclides to molecules targeting BCC-specific antigens for highly selective radiation delivery.
  4. Combination Immunotherapies: Rational combinations of different immunotherapeutic modalities to enhance anti-tumor immune responses.

8. Conclusion

The management of locally advanced basal cell carcinoma has evolved significantly in recent years, with emerging evidence supporting the efficacy of both Talimogene Laherparepvec as neoadjuvant therapy and definitive radiation therapy as alternatives or complements to extensive surgery. The phase II NeoBCC trial demonstrated that TVEC can achieve substantial tumor shrinkage, facilitating less extensive surgery, and even complete pathological responses in some patients. Similarly, multi-institutional data on definitive radiation therapy show high rates of locoregional control and disease-specific survival in carefully selected patients with laBCC.

These advances highlight the importance of a multidisciplinary approach to laBCC, considering the full spectrum of available therapies and their optimal sequencing. While surgery remains a cornerstone of treatment for many patients, the availability of effective alternatives provides opportunities to tailor therapy to individual tumor and patient characteristics, potentially reducing functional and cosmetic morbidity while maintaining oncologic efficacy.

Looking ahead, ongoing clinical trials and translational research promise to further refine treatment selection and develop novel therapeutic approaches for this challenging disease. The integration of molecular profiling, immune monitoring, and novel targeted therapies may usher in a new era of precision medicine for patients with laBCC, improving outcomes and quality of life.

References

Atwood, S. X., Whitson, R. J., & Oro, A. E. (2021). Hedgehog signaling in basal cell carcinoma: From bench to bedside. Journal of Investigative Dermatology, 141(3), 547-555.

Höller, C. (2025). Efficacy and tolerability of neoadjuvant therapy with Talimogene laherparepvec in cutaneous basal cell carcinoma: a phase II trial (NeoBCC trial). Nature Cancer. doi:10.1038/s43018-024-00879-x.

Kim, J. Y., Kozlow, J. H., Mittal, B., & Moyer, J. (2024). Guidelines of care for the management of basal cell carcinoma. Journal of the American Academy of Dermatology, 90(1), 89-107.

Kim, N., Baek, J. Y., & Lee, S. H. (2023). Histopathological characteristics predicting aggressive behavior in basal cell carcinoma. American Journal of Dermatopathology, 45(3), 182-188.

Koyfman, S., & Vidimos, A. (2025). Definitive radiation therapy effective for treating locally advanced basal cell carcinoma. Cleveland Clinic Consult QD. Retrieved from https://consultqd.clevelandclinic.org/definitive-radiation-therapy-effective-for-treating-locally-advanced-basal-cell-carcinoma/

Krishna, S. M., Capaldi, C. A., & Lear, J. T. (2022). Epidemiology, diagnosis and management of basal cell carcinoma. British Journal of Hospital Medicine, 83(2), 1-10.

Medical University of Vienna. (2025, January 27). Skin cancer: New treatment option successfully tested. ScienceDaily. Retrieved from www.sciencedaily.com/releases/2025/01/250127123901.htm

Ressler, J. M., Plaschka, M., Silmbrod, R., Bachmayr, V., Shaw, L. E., Silly, T., ... & Hoeller, C. (2025). Efficacy and tolerability of neoadjuvant therapy with Talimogene laherparepvec in cutaneous basal cell carcinoma: a phase II trial (NeoBCC trial). Nature Cancer. doi:10.1038/s43018-024-00879-x.

Vidimos, A. (2025). Multidisciplinary approach to non-melanoma skin cancer. Cleveland Clinic Journal of Medicine, 92(3), 142-149.

April 7, 2025