In a groundbreaking development within the field of metastatic breast cancer treatment, the results of the BRACELET-01 (PrECOG 0113) trial have been published, shedding new light on the potential synergy between chemotherapy and oncolytic virus immunotherapy. This Phase II randomized controlled trial evaluated the safety and efficacy of pelareorep, an oncolytic virus derived from a naturally occurring, non-pathogenic reovirus, when combined with the chemotherapy agent paclitaxel, both with and without the addition of the immune checkpoint inhibitor avelumab. The focus of this investigation was on patients with unresectable metastatic hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-negative (HER2-) breast cancer who had previously experienced disease progression despite endocrine therapy combined with cyclin-dependent kinase 4/6 inhibitors.
The trial enrolled 48 patients between June 2020 and June 2022, with a median age of 55 years, encompassing a population predominantly white, non-Hispanic, and postmenopausal. Participants were randomly assigned to one of three distinct treatment groups after a safety lead-in phase for the triple combination: paclitaxel alone, paclitaxel plus pelareorep, or paclitaxel combined with pelareorep and avelumab. Each participant continued the assigned treatment until disease progression or the emergence of intolerable adverse effects, while intensive monitoring for safety and tolerability was sustained throughout the study duration.
Pelareorep acts through two complementary mechanisms that are particularly relevant in oncologic immunotherapy. Intravenous administration of the virus initiates a potent stimulation of the host’s anti-tumor immune response by selectively infecting and lysing tumor cells, releasing tumor-associated antigens. This antigen release enhances immune recognition and primes cytotoxic T cells to better identify and attack neoplastic cells. Furthermore, pelareorep has been noted to modulate the tumor microenvironment by upregulating PD-L1 expression, a ligand that when targeted by checkpoint inhibitors like avelumab, can restore T-cell functionality dampened by tumor immune evasion strategies.
.adsslot_XhY2QvzSB9{width:728px !important;height:90px !important;}
@media(max-width:1199px){ .adsslot_XhY2QvzSB9{width:468px !important;height:60px !important;}
}
@media(max-width:767px){ .adsslot_XhY2QvzSB9{width:320px !important;height:50px !important;}
}
ADVERTISEMENT
The rationale for incorporating pelareorep alongside paclitaxel stems from preclinical and pooled clinical data suggesting that this combination may potentiate immune activation beyond chemotherapy’s cytotoxic effects. Specifically, data amalgamated from four prior trials involving over 500 patients across diverse solid tumors revealed an improvement in overall survival for HR+ HER2- metastatic breast cancer patients receiving the paclitaxel-pelareorep regimen compared to paclitaxel alone. This prelude was pivotal in shaping BRACELET-01’s design to test whether sensitization to checkpoint blockade via avelumab could further escalate therapeutic response rates.
Ultimately, the trial’s primary clinical endpoint, the objective response rate (ORR) at 16 weeks, demonstrated numerically higher activity for the combination of paclitaxel plus pelareorep (31%) compared to paclitaxel monotherapy (20%) and the triple regimen including avelumab (14%). Similarly, progression-free survival (PFS) was extended to a median of 12.1 months in the dual combination arm versus 6.4 months in the paclitaxel-only group. However, the addition of avelumab paradoxically resulted in a median PFS of only 5.8 months, accompanied by increased toxicity and a muted T-cell expansion, implying potential immunosuppressive effects or immune exhaustion at play when all three agents were combined.
Immunologically, a critical finding emerged in the form of T-cell clonal expansion, a hallmark of adaptive immune response vigor and a predictor of durable anti-cancer immunity. By the fourth treatment cycle, patients receiving paclitaxel plus pelareorep exhibited a pronounced proliferation of T-cell clones, suggesting the virus’ capability to reshape the immune landscape synergistically with chemotherapy. Conversely, paclitaxel alone failed to induce this effect, and the inclusion of avelumab appeared to blunt T-cell dynamics despite checkpoint blockade’s theoretical benefit in reversing immune suppression.
Safety profiles revealed increased adverse events within both combination arms compared to paclitaxel monotherapy, with the triple regimen being particularly challenging in terms of tolerability. The observed toxicity underscores the importance of meticulously balancing immune activation against immune-related adverse events, especially when manipulating multifaceted immunotherapeutic pathways. This cautionary finding highlights that more is not always better in combinatorial immuno-oncology and that mechanistic understanding must guide clinical development to optimize patient outcomes.
Clinicians and researchers have expressed a guarded optimism regarding pelareorep’s unique niche in breast cancer therapy, particularly within HR+ HER2- subtypes which have historically been refractory to immunotherapy approaches. The capacity of pelareorep to render ‘cold’ tumors immunologically ‘hot’ offers an innovative avenue towards overcoming therapeutic resistance, especially in the context of cytostatic endocrine therapies failing to control advanced disease. The BRACELET-01 findings provide a compelling rationale for further exploration into refined regimens and dosing strategies to harness the full potential of integrated immuno-viral and chemotherapeutic synergy.
Looking ahead, the investigative team recognizes that more nuanced biomarker studies are necessary to elucidate the underlying mechanisms driving differential responses and to identify patient subgroups most likely to benefit from pelareorep-based combination therapies. Additionally, longitudinal immune monitoring and exploration of tumor microenvironment modulation will be critical to advancing these approaches beyond early-phase clinical trials. The interplay between oncolytic virotherapy, chemotherapy, and immune checkpoint modulation represents a frontier with immense promise but also considerable complexity.
Diving deeper into mechanistic insights, pelareorep’s ability to infect and replicate selectively within tumor cells is known to induce immunogenic cell death characterized by the release of damage-associated molecular patterns (DAMPs) and type I interferons. This sets off a cascade of innate immune activation which bridges into adaptive immune processes, effectively transforming the tumor milieu into an in situ vaccine. The trial’s findings of augmented T-cell proliferation reinforce these mechanistic hypotheses, underscoring the translational significance of pelareorep as an immunomodulatory agent beyond its oncolytic property.
Despite the ambiguous outcomes of the triple regimen combining pembrolizumab, pelareorep, and paclitaxel, the BRACELET-01 study launches pivotal questions about the timing, sequencing, and dosing of multiplex immunotherapies. Immune checkpoint inhibitors may require a primed cytokine and cellular environment to achieve optimal efficacy, and premature or concurrent administration with agents inducing excessive inflammation may paradoxically induce immune dysfunction or tolerance. Future studies may be compelled to stratify patients based on immune biomarkers or explore staggered treatment schedules.
In summary, the BRACELET-01 trial represents a compelling stride toward refining treatment paradigms in metastatic HR+ HER2- breast cancer, a domain traditionally associated with limited immunotherapy success. The intriguing immunologic effects observed with the paclitaxel-pelareorep combination, accompanied by meaningful clinical activity, demand further investigation through larger, randomized studies equipped with comprehensive immune profiling. This pioneering work speaks to a broader paradigm shift in oncology — harnessing the intricate cross-talk between viral immunotherapy, chemotherapy, and immune checkpoints to transform treatment-resistant cancers into manageable or potentially curable diseases.
Subject of Research: People
Article Title: A Phase II Randomized Study of Paclitaxel Alone or Combined with Pelareorep with or without Avelumab in Metastatic Hormone Receptor–Positive Breast Cancer: The BRACELET-01/PrE0113 Study
News Publication Date: 16-May-2025
Web References:
– https://clinicaltrials.gov/search?term=pre-0113
– https://aacrjournals.org/clincancerres/article/doi/10.1158/1078-0432.CCR-24-2701/762447/A-Phase-II-Randomized-Study-of-Paclitaxel-Alone-or
References:
1. Clark AS, Zhao F, Klein P, Montero AJ, Falkson C, Krill-Jackson E et al. A Phase II Randomized Study of Paclitaxel Alone or Combined with Pelareorep with or without Avelumab in Metastatic Hormone Receptor–Positive Breast Cancer: The BRACELET-01/PrE0113 Study. Clin Cancer Res 2025; https://doi.org/10.1158/1078-0432.CCR-24-2701
2. Gutierrez AA, Reid C, Dzugalo A, Crawford M, Cheetham K, Parsi M, et al. Pooled Data Analysis of the Safety and Tolerability of Intravenous Pelareorep in Combination with Chemotherapy in 500+ Cancer Patients. Ann Oncol 2017; 28: v403–27; https://doi.org/10.1093/annonc/mdx376.056
Keywords: Breast cancer, Immunotherapy, Cancer immunotherapy, Combination therapies, Cancer research
Tags: BRACELET-01 trialchemotherapy for advanced breast cancerendocrine therapy resistant breast cancerHER2-negative breast cancer treatmentimmune checkpoint inhibitor avelumabmetastatic hormone receptor-positive breast cancernovel treatment strategies for breast canceroncolytic virus immunotherapypatient demographics in cancer trialspelareorep and paclitaxel combinationPhase II randomized controlled trialsafety and efficacy of pelareorep